1
|
Kovalyova Y, De Leon C, Krasowska-Zoladek A, Suon S, Wong J, Young S, Lee Heberling J, Price L, Berger R, Magliaro B, Cheng YS, Peier A, Rothman DM, Walji A, Smith S, Marcus J, Han X, Usenovic M. Promoting Secretion of Pathological Tau Species Using an Induced Proximity Platform That Engages the Autophagy Pathway. ACS Chem Neurosci 2025. [PMID: 40344401 DOI: 10.1021/acschemneuro.5c00161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2025] Open
Abstract
Intracellular accumulation of aberrantly phosphorylated aggregated tau protein can contribute to neuronal dysfunction associated with many neurodegenerative diseases. Thus, removing such tau species is an attractive therapeutic hypothesis for these diseases. Targeted protein degradation (TPD) strategies leveraging the autophagy-lysosome pathway (ALP) are promising approaches to decrease protein aggregates by designating them for degradation. Here, we developed a novel heterobifunctional molecule, MRL828, combining a tau pathology-binding ligand and modified guanine moiety based on the autophagy-targeting chimaera technology to selectively designate aggregated tau proteins for clearance via the ALP. Surprisingly, the MRL828-dependent decrease in intracellular tau aggregates was dependent on the autophagosome, but not the lysosome. MRL828 treatment led to autophagosome-dependent secretion of oligomeric and phosphorylated tau species, suggesting a reduction of intracellular tau aggregates via secretory autophagy rather than degradation via the ALP. This work highlights a novel mechanism of action (MOA) of an ALP-based heterobifunctional molecule and a potential new strategy for the cellular removal of proteins of interest.
Collapse
Affiliation(s)
| | - Cesar De Leon
- Chemical Biology, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | | | - Sokreine Suon
- Neuroscience, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Jacky Wong
- Neuroscience, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Seth Young
- Chemical Biology, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | | | - Laura Price
- Quantitative Biosciences, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Raphaëlle Berger
- Discovery Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Brian Magliaro
- Quantitative Biosciences, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Yu-Shan Cheng
- Quantitative Biosciences, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Andrea Peier
- Quantitative Biosciences, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Deborah M Rothman
- Chemical Biology, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Abbas Walji
- Discovery Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Sean Smith
- Neuroscience, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Jacob Marcus
- Neuroscience, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Xiaoqing Han
- Chemical Biology, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Marija Usenovic
- Neuroscience, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| |
Collapse
|
2
|
Foncea A, Franchini N, Tobar I, Thienel S, Retamal IN, Cancino GI, Cornejo F. Ptprd deficiency promotes tau hyperphosphorylation and impairs cognitive function in aged mice. Biol Res 2025; 58:26. [PMID: 40329347 PMCID: PMC12054186 DOI: 10.1186/s40659-025-00607-4] [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: 02/07/2025] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Tau phosphorylation is a tightly regulated process that ensures proper neuronal function. Indeed, hyperphosphorylation of tau closely contributes to neuronal dysfunction leading to neurodegenerative diseases, including tauopathies, which are characterized by excessive and aberrant tau phosphorylation and cognitive decline. Therefore, it is important to understand how to regulate its phosphorylation. In this regard, the protein tyrosine phosphatase receptor delta (PTPRD) has been genetically implicated in tau pathology in humans, but the mechanisms underlying its role in tau regulation remain unclear. This study investigates the impact of Ptprd deficiency on tau phosphorylation, cognitive function, neuroinflammation, and synaptic markers in aging mice. RESULTS Mice lacking Ptprd showed increased tau phosphorylation at multiple sites associated with its pathological aggregation. This effect was accompanied by the activation of the tau-related kinase Abl1, particularly in the hippocampus. Behavioral assessments revealed significant impairments in learning and memory, demonstrating the functional impact of these alterations. Moreover, Ptprd knockout mice showed increased microgliosis in both the entorhinal cortex and the hippocampus, suggesting a pro-inflammatory response. Furthermore, the synaptic protein PSD95 was also reduced in the cortex, indicating potential synaptic dysfunction. CONCLUSIONS The loss of Ptprd leads to increased tau phosphorylation, cognitive impairments, microgliosis, and synaptic alterations in older mice. Our findings also suggest that Ptprd plays a critical role in maintaining tau homeostasis through the Abl1 kinase. This indicates a new potential therapeutic approach for tauopathies, where PTPRD could serve a protective role against tau-related pathologies and may act as a key modulator in disease progression.
Collapse
Affiliation(s)
- Analía Foncea
- Centro de Biología Integrativa, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Nayhara Franchini
- Centro de Biología Integrativa, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Isidora Tobar
- Centro de Biología Integrativa, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Sebastián Thienel
- Centro de Biología Integrativa, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Ignacio N Retamal
- Centro de Oncología de Precisión, Escuela de Medicina, Facultad de Medicina y Ciencias de la Salud, Universidad Mayor, Santiago, Chile
| | - Gonzalo I Cancino
- Laboratorio de Neurobiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Francisca Cornejo
- Centro de Biología Integrativa, Facultad de Ciencias, Universidad Mayor, Santiago, Chile.
| |
Collapse
|
3
|
Bruqi K, Strappazzon F. NDP52 and its emerging role in pathogenesis. Cell Death Dis 2025; 16:359. [PMID: 40319017 PMCID: PMC12049512 DOI: 10.1038/s41419-025-07668-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 04/09/2025] [Accepted: 04/14/2025] [Indexed: 05/07/2025]
Abstract
Autophagy is a pro-survival process that regulates the degradation and renewal of cellular components, making it a crucial mechanism for cellular homeostasis. There are selective forms of autophagy that are specific to a number of substrates, such as pathogens (bacteria or viruses), protein aggregates or excess/damaged organelles. These processes involve as key players autophagy receptors, that link the cargo to be degraded to the autophagic machinery. Among them, NDP52 (also known as CALCOCO2) has been described to act as a "bridge" between the autophagy machinery and (1) damaged mitochondria in the mitophagy process; (2) pathogens during xenophagy or (3) proteins in the process of aggrephagy. The aim of this review is to summarize the major functions of NDP52, and to highlight the existence of two human NDP52 variants that have been described as risk or protective factors for Crohn's disease or Multiple Sclerosis and Alzheimer's disease patients, respectively. As these three diseases share common pathological features that lead to inflammation, such as mitochondria or gut microbiota dysfunctions, but also pathogenic infections, it seems clear that NDP52 could be a key player at the crossroad by acting indirectly on inflammation, and therefore a potential target for clinical applications and benefits.
Collapse
Affiliation(s)
- Krenare Bruqi
- Univ Lyon, Univ Lyon 1, CNRS, INSERM, Physiopathologie et Génétique du Neurone et du muscle, UMR5261, U1315, Institut Neuromyogène, Lyon, France
| | - Flavie Strappazzon
- Univ Lyon, Univ Lyon 1, CNRS, INSERM, Physiopathologie et Génétique du Neurone et du muscle, UMR5261, U1315, Institut Neuromyogène, Lyon, France.
| |
Collapse
|
4
|
Sharma V, Verma R, Singh TG. Targeting hypoxia-related pathobiology in Alzheimer's disease: strategies for prevention and treatment. Mol Biol Rep 2025; 52:416. [PMID: 40266407 DOI: 10.1007/s11033-025-10520-4] [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: 07/16/2024] [Accepted: 04/15/2025] [Indexed: 04/24/2025]
Abstract
INTRODUCTION Alzheimer's Disease (AD) is a neurodegenerative condition characterised by cognitive decline and memory impairment. Recent research highlights the important role of hypoxia, a state of insufficient oxygen availability, in exacerbating AD pathogenesis. MATERIALS AND METHODS Through the use of a number of different search engines like Scopus, PubMed, Bentham, and Elsevier databases, a literature review was carried out for investigating the role of hypoxia mediated pathobiology in AD. Only peerreviewed articles published in reputable journals in English language were included. Conversely, non-peer-reviewed articles, conference abstracts, and editorials were excluded, along with studies lacking experimental or clinical relevance or those unavailable in full text. CONCLUSION Hypoxia exacerbates core pathological features such as oxidative stress, neuroinflammation, mitochondrial dysfunction, amyloid-beta (Aβ) dysregulation, and hyperphosphorylation of tau protein. These interlinked mechanisms establish a self-perpetuating cycle of neuronal damage, accelerating disease progression. Addressing hypoxia as a modifiable risk factor offers potential for both prevention and treatment of AD. Exploring hypoxia and the HIF signalling pathway may help counteract the neuropathological and symptomatic effects of neurodegeneration.
Collapse
Affiliation(s)
- Veerta Sharma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Reet Verma
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| |
Collapse
|
5
|
Vanderlinden G, Vandenberghe R, Vandenbulcke M, Van Laere K. The Current Role of Tau PET Imaging in Neurodegeneration. Semin Nucl Med 2025:S0001-2998(25)00031-5. [PMID: 40263023 DOI: 10.1053/j.semnuclmed.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Accepted: 03/19/2025] [Indexed: 04/24/2025]
Abstract
Neurodegenerative tauopathies are characterized by the pathological hyperphosphorylation of tau proteins that subsequently form aggregates. Tau PET tracers with affinity to bind these pathological tau aggregates have been developed to measure disease progression and to support therapeutic drug development. In this review, we summarize the pathophysiology of tau throughout the range of neurodegenerative tauopathies. We outline the available first- and second-generation tau PET tracers, with a focus on new tau PET tracer developments, and discuss the quantification of tau PET images. Next, we summarize how tau PET relates to cerebrospinal fluid and plasma tau biomarkers. Finally, we review the current recommendations on the clinical use of tau PET versus fluid tau biomarkers in diagnosis, prognosis and treatment development.
Collapse
Affiliation(s)
- Greet Vanderlinden
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Rik Vandenberghe
- Department of Neurology, University Hospitals UZ Leuven, Leuven, Belgium; Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Mathieu Vandenbulcke
- Research Group Psychiatry, Leuven Brain Institute, KU Leuven, Leuven, Belgium; Department of Geriatric Psychiatry, University Hospitals UZ Leuven, Leuven, Belgium
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, Leuven Brain Institute, KU Leuven, Leuven, Belgium; Division of Nuclear Medicine, University Hospitals UZ Leuven, Leuven, Belgium.
| |
Collapse
|
6
|
Mirabelli M, Chiefari E, Arcidiacono B, Salatino A, Pascarella A, Morelli M, Credendino SC, Brunetti FS, Di Vito A, Greco A, Huin V, Nicoletti F, Pierantoni GM, Fedele M, Aguglia U, Foti DP, Brunetti A. HMGA1 deficiency: a pathogenic link between tau pathology and insulin resistance. EBioMedicine 2025; 115:105700. [PMID: 40233659 PMCID: PMC12019291 DOI: 10.1016/j.ebiom.2025.105700] [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: 11/08/2024] [Revised: 03/29/2025] [Accepted: 04/01/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Growing evidence links tau-related neurodegeneration with insulin resistance and type 2 diabetes (T2D), though the underlying mechanisms remain unclear. Our previous research identified HMGA1 as crucial for insulin receptor (INSR) expression, with defects in the HMGA1 gene associated with insulin resistance and T2D. Here, we explore HMGA1 deficiency as a potential contributor to tauopathies, such as Alzheimer's disease (AD), and its connection to insulin resistance. METHODS Immunoblot analyses, protein-DNA interaction studies, ChIP-qPCR, and reporter gene assays were conducted in human and mouse neuronal cell models. Tau immunohistochemistry, behavioural studies, and brain glucose metabolism were analysed in Hmga1-knockout mice. Additionally, a case-control study investigated the relationship between HMGA1 and tau pathology in patients with tauopathy, carrying or not the HMGA1 rs146052672 variant, known to reduce HMGA1 protein levels and increase the risk of insulin resistance and T2D. FINDINGS We show that HMGA1 regulates tau protein expression primarily through the specific repression of MAPT gene transcription. In both human neuronal cells and primary mouse neurons, tau mRNA and protein levels were inversely correlated with HMGA1 expression. This inverse relationship was further confirmed in the brain of Hmga1-knockout mice, where tau was overexpressed, INSR was downregulated, and brain glucose uptake was impaired. Additionally, the rs146052672 variant was more common in patients with tauopathy (12/69, 17.4%) than in controls (10/200, 5.0%) (p = 0.001), and carriers of this variant exhibited more severe disease progression and poorer therapeutic outcomes. INTERPRETATION These findings suggest that HMGA1 deficiency may drive tau pathology, linking tauopathies to insulin resistance and providing new insights into the relationship between metabolic and neurodegenerative disorders. Furthermore, our observation that over 17% of individuals with tauopathy exhibit a deficit in HMGA1 protein production could have significant clinical implications, potentially guiding the development of therapeutic strategies targeting this specific defect. FUNDING See acknowledgements section.
Collapse
Affiliation(s)
- Maria Mirabelli
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Eusebio Chiefari
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Biagio Arcidiacono
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Alessandro Salatino
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Angelo Pascarella
- Department of Medical and Surgical Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Maurizio Morelli
- Department of Medical and Surgical Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Sara C Credendino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Francesco S Brunetti
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Anna Di Vito
- Department of Experimental and Clinical Medicine, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Adelaide Greco
- Interdepartmental Centre of Veterinary Radiology, University of Naples "Federico II", Naples, Italy
| | - Vincent Huin
- University of Lille, Inserm, CHU-Lille, Lille Neuroscience & Cognition, UMR-S1172, Team Alzheimer & Tauopathies, F-59000, Lille, France
| | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology, University of Rome "Sapienza", Rome, Italy; IRCCS Neuromed, Pozzilli, Italy
| | - Giovanna M Pierantoni
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Monica Fedele
- Institute of Experimental Endocrinology and Oncology, CNR, Naples, Italy
| | - Umberto Aguglia
- Department of Medical and Surgical Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy
| | - Daniela P Foti
- Department of Experimental and Clinical Medicine, University of Catanzaro "Magna Græcia", Catanzaro, Italy.
| | - Antonio Brunetti
- Department of Health Sciences, University of Catanzaro "Magna Græcia", Catanzaro, Italy.
| |
Collapse
|
7
|
Tanabe H, Maeda S, Sano E, Sakai N, Endoh-Yamagami S, Okano H. Tau aggregation induces cell death in iPSC-derived neurons. AGING BRAIN 2025; 7:100136. [PMID: 40276591 PMCID: PMC12018045 DOI: 10.1016/j.nbas.2025.100136] [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: 12/08/2024] [Revised: 03/31/2025] [Accepted: 04/04/2025] [Indexed: 04/26/2025] Open
Abstract
Abnormal accumulation of tau proteins in the brain is a hallmark of neurodegenerative diseases such as Alzheimer's disease and is closely linked with neuronal cell death. Tau accumulation is a prominent therapeutic target for Alzheimer's disease, since tau accumulation correlates well with the disease progression, and tau-targeting drugs hold potentials to halt the disease progression. Given the differential response of human and mouse neuronal cells, there is a critical need for a human cellular platform to quickly screen for tau-related neurodegenerative disease therapeutics. However, inducing rapid, tau-dependent neuronal cell death in human models remains challenging. In this study, we established a human cellular model capable of inducing tau aggregation-dependent neuronal cell death within two weeks via tau overexpression. Additionally, we demonstrated the neuroprotective efficacy of known tau-targeting compounds within this system. These findings suggest that our cellular model recapitulates the molecular pathogenesis of tau-induced neurodegeneration and could serve as a valuable platform for drug screening in tauopathies.
Collapse
Affiliation(s)
- Hirokazu Tanabe
- FUJIFILM Corporation, Bio Science & Engineering Laboratories, Kanagawa, Japan
| | - Sumihiro Maeda
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Etsuko Sano
- Keio University Regenerative Medicine Research Center, Kanagawa, Japan
| | - Norio Sakai
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical & Health Sciences Hiroshima University, Hiroshima, Japan
| | | | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Keio University Regenerative Medicine Research Center, Kanagawa, Japan
| |
Collapse
|
8
|
Rahman MW, Sharma P, Chattopadhyay T, Saroja SR. Distinct neuronal vulnerability and metabolic dysfunctions are characteristic features of fast-progressing Alzheimer's patients with Lewy bodies. J Biol Chem 2025; 301:108396. [PMID: 40074078 PMCID: PMC12002820 DOI: 10.1016/j.jbc.2025.108396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 02/12/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
Tau protein accumulation is linked to dementia progression in Alzheimer's disease (AD), with potential co-pathologies contributing to it. The progression of dementia in patients with AD varies between individuals, and the association between co-pathology and heterogeneity in dementia progression rate remains unclear. We used longitudinal cohort data, postmortem brain tissues, and biochemical methods such as immunoassays and proteomic profiling to investigate the molecular components associated with progression rate. We report that AD with comorbidities, such as dementia with Lewy bodies (DLB) and TDP-43 pathology, progress faster than AD alone. Patients with AD-DLB had higher levels of soluble oligomeric tau proteins and lower levels of insoluble tau proteins compared to those with AD alone. Our data suggest that α-synuclein fibrils may enhance tau aggregation through cross-seeding. The prefrontal cortex is more vulnerable to Lewy body pathology than the temporal cortex, and Tau and α-synuclein aggregate in distinct neuronal populations, indicating selective neuronal and regional vulnerability to their respective pathologies. Dysfunctional metabolic pathways were more strongly associated with patients having fast-progressing AD-DLB. Our study suggests that comorbidities, such as α-synuclein aggregation and metabolic dysfunctions, are associated with rapidly progressing AD patients, highlighting the importance of patient subgrouping for clinical trials.
Collapse
Affiliation(s)
- Mohammed Waseequr Rahman
- Center for Brain Research, Indian Institute of Science, Bangalore, India; Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Preeti Sharma
- Center for Brain Research, Indian Institute of Science, Bangalore, India; Manipal Academy of Higher Education, Manipal, Karnataka, India
| | | | | |
Collapse
|
9
|
Wu Y, Li T, Jiang X, Ling J, Zhao Z, Zhu J, Chen C, Liu Q, Yang X, Shen X, Ma R, Li G, Liu G. (-)-Epicatechin Rescues Memory Deficits by Activation of Autophagy in a Mouse Model of Tauopathies. MedComm (Beijing) 2025; 6:e70144. [PMID: 40135197 PMCID: PMC11933444 DOI: 10.1002/mco2.70144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 01/23/2025] [Accepted: 02/13/2025] [Indexed: 03/27/2025] Open
Abstract
In tauopathies, defects in autophagy-lysosomal protein degradation are thought to contribute to the abnormal accumulation of aggregated tau. Recent studies have shown that (-)-Epicatechin (Epi), a dietary flavonoid belonging to the flavan-3-ol subgroup, improves blood flow, modulates metabolic profiles, and prevents oxidative damage. However, less research has explored the effects of Epi on tauopathies. Here, we found that Epi rescued cognitive deficits in P301S tau transgenic mice, a model exhibiting characteristics of tauopathies like frontotemporal dementia and Alzheimer's disease, and attenuated tau pathology through autophagy activation. Proteomic and biochemical analyses revealed that P301S mice exhibit deficits in autophagosome formation via modulating mTOR, consequently inhibiting autophagy. Epi inhibited the mTOR signaling pathway to promote autophagosome formation, which is essential for the clearance of tau aggregation. By using chloroquine (CQ) to inhibit autophagy in vivo, we further confirmed that Epi induced tau degradation via the autophagy pathway. Lastly, Epi administration was also found to improve cognition by reversing spine decrease and neuron loss, as well as attenuating neuroinflammation. Our findings suggest that Epi promoted tau clearance by activating autophagy, indicating its potential as a promising therapeutic candidate for tauopathies.
Collapse
Affiliation(s)
- Yanqing Wu
- Department of NeurologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Health Management CenterRenmin Hospital of Wuhan UniversityWuhanChina
| | - Ting Li
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Ministry of Education of China and Hubei Province for Neurological DisordersTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of PathologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Xingjun Jiang
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jianmin Ling
- Department of Emergency MedicineTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Critical Care MedicineTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Zaihua Zhao
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational EnvironmentSchool of Public HealthAir Force Medical UniversityXi'anChina
| | - Jiahui Zhu
- Department of NeurologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Chongyang Chen
- Key Laboratory of Nuclear MedicineMinistry of HealthJiangsu Key Laboratory of Molecular Nuclear MedicineJiangsu Institute of Nuclear MedicineWuxiChina
| | - Qian Liu
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Ministry of Education of China and Hubei Province for Neurological DisordersTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of ShenzhenShenzhen Center for Disease Control and PreventionShenzhenChina
| | - Xuefeng Shen
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational EnvironmentSchool of Public HealthAir Force Medical UniversityXi'anChina
| | - Rong Ma
- Department of PharmacologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Gang Li
- Department of NeurologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Gongping Liu
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Ministry of Education of China and Hubei Province for Neurological DisordersTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Modern Toxicology of ShenzhenShenzhen Center for Disease Control and PreventionShenzhenChina
- Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| |
Collapse
|
10
|
Tang J, Xu Z, Wang F, Guan L, Qi B, Zou Y. Caffeine Inhibits Tau Aggregation and Destabilizes the Fibril Associated with Chronic Traumatic Encephalopathy: A REST2 and Conventional MD Simulations Study. J Chem Inf Model 2025; 65:2985-2998. [PMID: 40053114 DOI: 10.1021/acs.jcim.4c02353] [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: 03/25/2025]
Abstract
Chronic traumatic encephalopathy (CTE) is a unique tauopathy mostly diagnosed in contact sports athletes, such as those active in American football, boxing, soccer, etc. The hyperphosphorylated fibrillar aggregates composed of self-assembled tau protein are the pathological hallmark of CTE, and inhibiting the aggregation or disassociating the fibrillar aggregates has been considered a promising avenue to prevent or treat CTE. Caffeine (CA) is a well-known psychostimulant and can be found in coffee, tea, and soft drinks. In vitro experiments revealed that CA could effectively inhibit wild-type tau aggregation and disassemble preformed fibrils. However, the atomic effect and the underlying molecular mechanisms remain largely elusive. In this study, we performed a multitude of replica exchange with solute tempering 2 (REST2) and conventional molecular dynamics (CMD) simulations of 43.8 μs in total on tau models with and without CA, including the third and fourth microtubule-binding repeats (R3-R4) tau monomer and CTE-related R3-R4 tau protofibril and fibril. The results revealed that CA could prominently inhibit the β-sheet formation of the monomer and disrupt the β-sheet structure of the protofibril, inducing the monomer and protofibril to adopt loosely packed or extended conformations. H-bonding and π-π stacking interactions drove the binding of CA on the monomer, while hydrophobic interactions made an extra contribution to the binding of CA on the protofibril. Strikingly, CA could stably bind to the hydrophobic cavity of the protofibril, which might occupy the space and prevent the entering of the aggregation cofactor. What is more, CA destabilized the fibril and played a role in reversing the liquid-to-solid phase transition (LSPT) of tau. Our study systematically uncovered the atomic-level effect of CA on tau aggregation, which offers a theoretical foundation for the design of drugs to prevent or treat CTE.
Collapse
Affiliation(s)
- Jiaxing Tang
- School of Physical Education, Xiangnan University, 889 Chenzhou Avenue, Chenzhou 423000, People's Republic of China
| | - Zhengdong Xu
- Department of Physical Education, Shanghai University of Engineering Science, 333 Long Teng Road, Shanghai 201620, People's Republic of China
| | - Feng Wang
- School of Physical Education, Xiangnan University, 889 Chenzhou Avenue, Chenzhou 423000, People's Republic of China
| | - Lulu Guan
- Department of Sport and Exercise Science, College of Education, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, People's Republic of China
| | - Bote Qi
- Department of Sport and Exercise Science, College of Education, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, People's Republic of China
| | - Yu Zou
- Department of Sport and Exercise Science, College of Education, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, Zhejiang, People's Republic of China
| |
Collapse
|
11
|
Zaman M, Yang S, Huang Y, Yarbro JM, Hao Y, Wang Z, Liu D, Harper KE, Soliman H, Hemphill A, Harvey S, Pruett-Miller SM, Stewart V, Tanwar AS, Kalathur R, Grace CR, Turk M, Chittori S, Jiao Y, Wu Z, High AA, Wang X, Serrano GE, Beach TG, Yu G, Yang Y, Chen PC, Peng J. Midkine Attenuates Aβ Fibril Assembly and Amyloid Plaque Formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.20.644383. [PMID: 40166321 PMCID: PMC11957132 DOI: 10.1101/2025.03.20.644383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Proteomic profiling of Alzheimer's disease (AD) brains has identified numerous understudied proteins, including midkine (MDK), that are highly upregulated and correlated with Aβ since the early disease stage, but their roles in disease progression are not fully understood. Here we present that MDK attenuates Aβ assembly and influences amyloid formation in the 5xFAD amyloidosis mouse model. MDK protein mitigates fibril formation of both Aβ40 and Aβ42 peptides in Thioflavin T fluorescence assay, circular dichroism, negative stain electron microscopy, and NMR analysis. Knockout of Mdk gene in 5xFAD increases amyloid formation and microglial activation. Further comprehensive mass spectrometry-based profiling of whole proteome and detergent-insoluble proteome in these mouse models indicates significant accumulation of Aβ and Aβ-correlated proteins, along with microglial components. Thus, our structural and mouse model studies reveal a protective role of MDK in counteracting amyloid pathology in Alzheimer's disease.
Collapse
Affiliation(s)
- Masihuz Zaman
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Shu Yang
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Present address: Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, P. R. China
| | - Ya Huang
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Jay M. Yarbro
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Yanhong Hao
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Zhen Wang
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Danting Liu
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Kiara E. Harper
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Hadeer Soliman
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Alex Hemphill
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Sarah Harvey
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Shondra M. Pruett-Miller
- Center for Advanced Genome Engineering, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Valerie Stewart
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Ajay Singh Tanwar
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Ravi Kalathur
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Christy R. Grace
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Martin Turk
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Sagar Chittori
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Yun Jiao
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Zhiping Wu
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Anthony A. High
- Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Xusheng Wang
- Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | | | - Thomas G. Beach
- Banner Sun Health Research Institute, Sun City, AZ 85351, USA
| | - Gang Yu
- Department of Neuroscience, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yang Yang
- Department of Structural Biology, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Ping-Chung Chen
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Junmin Peng
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
12
|
Langer Horvat L, Španić Popovački E, Babić Leko M, Zubčić K, Mustapić M, Hof PR, Šimić G. Biochemical characterization of Tau protein changes and amyloid dynamics in a novel non-transgenic rat model of tauopathy. J Neural Transm (Vienna) 2025:10.1007/s00702-025-02909-z. [PMID: 40095078 DOI: 10.1007/s00702-025-02909-z] [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: 01/18/2025] [Accepted: 03/04/2025] [Indexed: 03/19/2025]
Abstract
In this study, we further characterized a non-transgenic model of tauopathy by examining tau protein changes using ELISA and Western blot upon inoculation of human tau oligomers (TO) and human tau synthetic pre-formed fibrils (TF) into the medial entorhinal cortex of Wistar rats. Our analyses showed that inoculation with TO did not significantly alter the ratio of phosphorylated tau at AT8 epitopes (pSer202/pThr205) to total tau protein in the hippocampus and entorhinal cortex, but only resulted in a decrease of phosphorylation at AT100 epitopes (pThr212/pSer214). As we previously observed an increase in AT8 immunostaining in both regions, this suggests method-dependent conformational alterations. In contrast, eleven months after inoculation, TF caused significant AT8 and PHF-1 (pSer396/pSer404) epitope-specific changes in tau phosphorylation in the hippocampus, but not in the entorhinal cortex, reflecting a more advanced stage of Alzheimer's disease (AD)-like changes compared to TO. Importantly, amyloid plaques appeared as early as four months post-inoculation with TO, preceding significant phosphorylation changes of tau, thus indicating that amyloid probably facilitates early tau seeding and spreading. This was corroborated by the observed dynamic changes in Aβ1-42 levels in cerebrospinal fluid, with initial decreases followed by increases, similar to patterns seen in transgenic mouse models of AD and in AD patients. Altogether, these findings lead us to conclude that changes in tau protein induce amyloid changes and vice versa, which is actually what defines AD as a unique neurodegenerative disease.
Collapse
Affiliation(s)
- Lea Langer Horvat
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, 10000, Croatia
| | - Ena Španić Popovački
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, 10000, Croatia
| | - Mirjana Babić Leko
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, 10000, Croatia
- Department of Medical Biology, University of Split Faculty of Science, Split, 21000, Croatia
| | - Klara Zubčić
- Department of Laboratory Diagnostics, Dubrava University Hospital, Zagreb, 10000, Croatia
| | - Maja Mustapić
- Laboratory of Clinical Investigation, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Patrick R Hof
- Nash Family Department of Neuroscience, Friedman Brain Institute, and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Goran Šimić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, 10000, Croatia.
| |
Collapse
|
13
|
Kahil E, Jackson A, D'Souza B, Gorman G, Jumbo-Lucioni P. Investigating the therapeutic potential of 1,2,3,4,6 penta-O-galloyl-β-D-glucose in Alzheimer's disease: a scoping review. Nat Prod Res 2025:1-15. [PMID: 40096746 DOI: 10.1080/14786419.2025.2477226] [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: 10/12/2024] [Accepted: 03/03/2025] [Indexed: 03/19/2025]
Abstract
The gallotannin 1,2,3,4,6-Penta-O-Galloyl-β-D-Glucose (β-PGG) has recently garnered scientific interest due to its potent antioxidant, anti-inflammatory and neuroprotective properties. Alzheimer's disease (AD), a neurodegenerative disorder marked by neuroinflammation, oxidative stress, cholinergic dysfunction, accumulation of amyloid β (Aβ), tau and metal ions, may benefit from β-PGG interventions. This review synthesises existing literature to explore the therapeutic potential of β-PGG in AD. Experimental evidence suggests that β-PGG can suppress pro-inflammatory mediator release and inhibit NF-κB and MAPK signalling pathways, both critical in AD progression. In vitro studies highlight β-PGG's ability to scavenge free radicals, enhance antioxidant enzyme function, and prevent lipid peroxidation, well-known phenomena in AD. Additionally, neuroprotection may be conferred by β-PGG's capacity to reduce metal ion accumulation, inhibit Aβ and tau aggregation, and modulate cholinergic neurotransmission in vitro. In summary, while β-PGG shows promise in targeting AD hallmarks in vitro, further in vivo studies are crucial to validate its therapeutic potential.
Collapse
Affiliation(s)
- Ezaldean Kahil
- McWhorter School of Pharmacy, Samford University, Birmingham, Alabama, USA
| | - Anisha Jackson
- McWhorter School of Pharmacy, Samford University, Birmingham, Alabama, USA
| | - Bernadette D'Souza
- Department of Pharmaceutical, Social and Administrative Sciences, McWhorter School of Pharmacy, Samford University, Birmingham, Alabama, USA
| | - Gregory Gorman
- Department of Pharmaceutical, Social and Administrative Sciences, McWhorter School of Pharmacy, Samford University, Birmingham, Alabama, USA
- Pharmaceutical Sciences Research Institute, Samford University, Birmingham, Alabama, USA
| | - Patricia Jumbo-Lucioni
- Department of Pharmaceutical, Social and Administrative Sciences, McWhorter School of Pharmacy, Samford University, Birmingham, Alabama, USA
- Department of Biology, College of Arts and Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
14
|
Azam U, Naseer MM, Rochais C. Analysis of skeletal diversity of multi-target directed ligands (MTDLs) targeting Alzheimer's disease. Eur J Med Chem 2025; 286:117277. [PMID: 39848035 DOI: 10.1016/j.ejmech.2025.117277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/10/2025] [Accepted: 01/11/2025] [Indexed: 01/25/2025]
Abstract
Alzheimer's disease (AD) remains a significant healthcare challenge, necessitating innovative therapeutic approaches to address its complex and multifactorial nature. Traditional drug discovery strategies targeting single molecular targets are not sufficient for the effective treatment of AD. In recent years, MTDLs have emerged as promising candidates for AD therapy, aiming to simultaneously modulate multiple pathological targets. Among the various strategies employed in MTDL design, pharmacophore hybridization offers a versatile approach to integrate diverse pharmacophoric features within a single molecular scaffold. This strategy provides access to a wide array of chemical space for the design and development of novel therapeutic agents. This review, therefore, provides a comprehensive overview of skeletal diversity exhibited by MTDLs designed recently for AD therapy based on pharmacophore hybridization approach. A diverse range of pharmacophoric elements and core scaffolds hybridized to construct MTDLs that has the potential to target multiple pathological features of AD including amyloid-beta aggregation, tau protein hyperphosphorylation, cholinergic dysfunction, oxidative stress, and neuroinflammation are discussed. Through the comprehensive analysis and integration of structural insights of key biomolecular targets, this review aims to enhance optimization efforts in MTDL design, ultimately striving towards a comprehensive cure for the multifaceted pathophysiology of the disease.
Collapse
Affiliation(s)
- Uzma Azam
- Department of Chemistry, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Muhammad Moazzam Naseer
- Department of Chemistry, Quaid-i-Azam University, Islamabad, 45320, Pakistan; Université de Caen Normandie, Normandie Univ., CERMN, 14000, Caen, France.
| | - Christophe Rochais
- Université de Caen Normandie, Normandie Univ., CERMN, 14000, Caen, France.
| |
Collapse
|
15
|
Mu J, Zhang Z, Jiang C, Geng H, Duan J. Role of Tau Protein Hyperphosphorylation in Diabetic Retinal Neurodegeneration. J Ophthalmol 2025; 2025:3278794. [PMID: 40109357 PMCID: PMC11922625 DOI: 10.1155/joph/3278794] [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: 01/05/2024] [Revised: 12/25/2024] [Accepted: 02/22/2025] [Indexed: 03/22/2025] Open
Abstract
Diabetic retinal neurodegeneration (DRN) is an early manifestation of diabetic retinopathy (DR) characterized by neurodegeneration that precedes microvascular abnormalities in the retina. DRN is characterized by apoptosis of retinal ganglion cells (involves alterations in retinal ganglion cells [RGCs], photoreceptors, amacrine cells and bipolar cells and so on), reactive gliosis, and reduced retinal neuronal function. Tau, a microtubule-associated protein, is a key mediator of neurotoxicity in neurodegenerative diseases, with functions in phosphorylation-dependent microtubule assembly and stabilization, axonal transport, and neurite outgrowth. The hyperphosphorylated tau (p-tau) loses its ability to bind to microtubules and aggregates to form paired helical filaments (PHFs), which further form neurofibrillary tangles (NFTs), leading to abnormal cell scaffolding and cell death. Studies have shown that p-tau can cause degeneration of RGCs in DR, making tau pathology a new pathophysiological model for DR. Here, we review the mechanisms by which p-tau contribute to DRN, including insulin resistance or lack of insulin, mitochondrial damage such as mitophagy impairment, mitochondrial axonal transport defects, mitochondrial bioenergetics dysfunction, and impaired mitochondrial dynamics, Abeta toxicity, and inflammation. Therefore, this article proposes that tau protein hyperphosphorylation plays a crucial role in the pathogenesis of DRN and may serve as a novel therapeutic target for combating DRN.
Collapse
Affiliation(s)
- Jingyu Mu
- Eye School of Chengdu University of TCM, Chengdu, Sichuan, China
- Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM Laboratory, Chengdu, Sichuan, China
- Retinal Image Technology and Chronic Vascular Disease Prevention & Control and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Zengrui Zhang
- Eye School of Chengdu University of TCM, Chengdu, Sichuan, China
- Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM Laboratory, Chengdu, Sichuan, China
- Retinal Image Technology and Chronic Vascular Disease Prevention & Control and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Chao Jiang
- College of Life and Health Sciences, Institute of Neuroscience, Northeastern University, Shenyang, China
| | - Haoming Geng
- Eye School of Chengdu University of TCM, Chengdu, Sichuan, China
- Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM Laboratory, Chengdu, Sichuan, China
- Retinal Image Technology and Chronic Vascular Disease Prevention & Control and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Junguo Duan
- Eye School of Chengdu University of TCM, Chengdu, Sichuan, China
- Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM Laboratory, Chengdu, Sichuan, China
- Retinal Image Technology and Chronic Vascular Disease Prevention & Control and Collaborative Innovation Center, Chengdu, Sichuan, China
- Ineye Hospital of Chengdu University of TCM, Chengdu, Sichuan, China
| |
Collapse
|
16
|
Tsoi PS, Lucas L, Rhoades D, Ferreon JC, Ferreon ACM. Electrostatic Effects on Tau Nanocondensates. Biomolecules 2025; 15:406. [PMID: 40149942 PMCID: PMC11940141 DOI: 10.3390/biom15030406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Biomolecular condensates (BMCs) are membrane-less protein compartments with physiological and pathological relevance. The formation of BMCs is driven by a process known as liquid-liquid phase separation (LLPS), a field that has largely focused on the study of micron-sized condensates. However, there have been recent studies showing that proteins that undergo LLPS also form nanometer-sized condensates. These nanometer-sized condensates, or nanocondensates, are distinct from microcondensates and potentially exhibit more relevance in cell biology. The field of nanocondensate research is in its infancy, with limited biophysical studies of these structures. Here, we studied condensate formation and dissolution of wild-type and disease-linked (hyperphosphorylated and missense mutated) Tau. We investigated the effects of solution condition modulation on nanocondensate formation and dissolution, and observed that Tau condensation is strongly regulated by electrostatic forces and less affected by hydrophobic disruption. We observed that all three Tau variants studied shared condensate formation properties when in solution conditions with the same ionic strength. However, hyperphosphorylated and missense-mutated Tau exhibited higher resistance to dissolution compared to wild-type Tau. This study uncovers additional distinctions between different types of condensates, which provides further insight into the distinctions between physiological and pathological condensates.
Collapse
Affiliation(s)
- Phoebe S. Tsoi
- Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA; (P.S.T.); (L.L.)
| | - Lathan Lucas
- Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA; (P.S.T.); (L.L.)
| | - Derek Rhoades
- Department of Chemistry, Scripps Research, La Jolla, CA 92037, USA;
| | - Josephine C. Ferreon
- Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA; (P.S.T.); (L.L.)
| | - Allan Chris M. Ferreon
- Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX 77030, USA; (P.S.T.); (L.L.)
| |
Collapse
|
17
|
Chakravarti A, Joseph JA. Accurate prediction of thermoresponsive phase behavior of disordered proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.04.641540. [PMID: 40093057 PMCID: PMC11908177 DOI: 10.1101/2025.03.04.641540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Protein responses to environmental stress, particularly temperature fluctuations, have long been a subject of investigation, with a focus on how proteins maintain homeostasis and exhibit thermoresponsive properties. While UCST-type (upper critical solution temperature) phase behavior has been studied extensively and can now be predicted reliably using computational models, LCST-type (lower critical solution temperature) phase transitions remain less explored, with a lack of computational models capable of accurate prediction. This gap limits our ability to probe fully how proteins undergo phase transitions in response to temperature changes. Here, we introduce Mpipi-T, a residue-level coarse-grained model designed to predict LCST-type phase behavior of proteins. Parametrized using both atomistic simulations and experimental data, Mpipi-T accounts for entropically driven protein phase separation that occurs upon heating. Accordingly, Mpipi-T predicts temperature-driven protein behavior quantitatively in both single- and multi-chain systems. Beyond its predictive capabilities, we demonstrate that Mpipi-T provides a framework for uncovering the molecular mechanisms underlying heat stress responses, offering new insights into how proteins sense and adapt to thermal changes in biological systems.
Collapse
Affiliation(s)
- Ananya Chakravarti
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
- Omenn–Darling Bioengineering Institute, Princeton University, Princeton, NJ 08544, USA
| | - Jerelle A. Joseph
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA
- Omenn–Darling Bioengineering Institute, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
18
|
Acosta K, Brue CR, Holubovska P, Kim HJ, Mayne L, Murakami K, Rhoades E. Structural insights into the role of the proline rich region in tau function. Structure 2025; 33:465-474.e8. [PMID: 39826549 PMCID: PMC11890945 DOI: 10.1016/j.str.2024.12.017] [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: 02/14/2024] [Revised: 09/16/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025]
Abstract
Tau plays an important role in modulating axonal microtubules in neurons, while intracellular tau aggregates are found in many neurodegenerative disorders. Tubulin binding sites are found in tau's proline-rich region (PRR), microtubule binding repeats (MTBRs), and pseudo-repeat (R'). Tau phosphorylation sites, which cluster with high frequency within the PRR, regulate tubulin interactions and correlates with disease. Here, we use fluorescence correlation spectroscopy and structural mass spectrometry techniques to characterize the impact of phosphomimic mutations in the PRR on tau function. We find that phosphomimics cumulatively diminish tubulin dimer binding and slow microtubule polymerization. Additionally, we map two ∼15 residue regions of the PRR as primary tubulin dimer binding sites and propose a model in which PRR enhances lateral interactions between tubulin dimers, complementing the longitudinal interactions observed for MTBR. Our study provides insight into the previously overlooked relevance of tau's PRR in functional interactions with tubulin dimers.
Collapse
Affiliation(s)
- Karen Acosta
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christopher R Brue
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Polina Holubovska
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hee Jong Kim
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Leland Mayne
- Johnson Research Foundation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19355, USA; Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kenji Murakami
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elizabeth Rhoades
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Chemistry, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
19
|
Alten B, Gokcal E, Warren A, van Veluw SJ, Kozberg M, Gurol ME, Viswanathan A, Greenberg SM. Cerebrospinal Fluid Beta-Amyloid Concentration and Clinical and Radiographic Manifestations of Cerebral Amyloid Angiopathy. J Am Heart Assoc 2025; 14:e040025. [PMID: 40028849 DOI: 10.1161/jaha.124.040025] [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: 11/20/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Cerebral amyloid angiopathy (CAA) is driven by vascular Aβ (amyloid-beta) deposition, which can be detected as reduced Aβ species in cerebrospinal fluid (CSF). We sought to identify relationships between CSF Aβ and tau concentrations and various manifestations of CAA. METHODS This is a retrospective cross-sectional single-center study of patients diagnosed with CAA per Boston Criteria version 2.0, had magnetic resonance imaging brain scans, and underwent CSF testing for Aβ and tau concentrations between 2008 and 2022. Associations between clinical/magnetic resonance imaging features and CSF biomarker concentrations were investigated with univariate and multivariate models. RESULTS We identified 31 patients aged 69.6±8.4 years, of whom 20 presented with cognitive complaints, 9 with CAA-related macrohemorrhage (lobar intraparenchymal or convexity subarachnoid hemorrhage), and 2 with transient focal neurological episodes. Presence of macrohemorrhage (301.8±112 pg/mL versus 400.9±123 pg/mL, P=0.029), cortical superficial siderosis (309.6±131 mg/dL versus 412.3±100 pg/mL, P=0.021), and severe enlarged perivascular spaces in centrum semiovale (285.8±91 pg/mL versus 428.3±117 pg/mL, P<0.001) were associated with lower Aβ42 concentrations. Aβ42 concentrations inversely correlated with the number of these manifestations, being lowest in patients having all three. Patients with cognitive complaints had higher t-tau (total tau; 551±320 pg/mL versus 317.2±141 pg/mL, P=0.03) and trended toward having higher p-tau (phosphorylated tau at threonine 181) concentrations (75.69±39 pg/mL versus 49.24±22 pg/mL, P=0.05). CONCLUSIONS Lower CSF Aβ42, suggesting higher amyloid burden, is associated with CAA-related macrohemorrhages and severe enlarged perivascular spaces in centrum semiovale, suggesting potential mechanistic links and CSF Aβ42 as a potential biomarker for progression of CAA. CSF tau concentrations are linked to cognitive complaints, likely representing comorbid Alzheimer disease pathology.
Collapse
Affiliation(s)
- Baris Alten
- Mass General Brigham Neurology Residency Program Boston MA USA
- Department of Neurology Massachusetts General Hospital Boston MA USA
- Department of Neurology Brigham and Women's Hospital Boston MA USA
| | - Elif Gokcal
- Department of Neurology Massachusetts General Hospital Boston MA USA
| | - Andrew Warren
- Department of Neurology Massachusetts General Hospital Boston MA USA
| | | | - Mariel Kozberg
- Department of Neurology Massachusetts General Hospital Boston MA USA
| | - M Edip Gurol
- Department of Neurology Massachusetts General Hospital Boston MA USA
| | - Anand Viswanathan
- Department of Neurology Massachusetts General Hospital Boston MA USA
| | | |
Collapse
|
20
|
Schaible P, Henschel J, Erny D. How the gut microbiota impacts neurodegenerative diseases by modulating CNS immune cells. J Neuroinflammation 2025; 22:60. [PMID: 40033338 PMCID: PMC11877772 DOI: 10.1186/s12974-025-03371-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/06/2025] [Indexed: 03/05/2025] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease worldwide. Amyloid-β (Aβ) accumulation and neurofibrillary tangles are two key histological features resulting in progressive and irreversible neuronal loss and cognitive decline. The macrophages of the central nervous system (CNS) belong to the innate immune system and comprise parenchymal microglia and CNS-associated macrophages (CAMs) at the CNS interfaces (leptomeninges, perivascular space and choroid plexus). Microglia and CAMs have received attention as they may play a key role in disease onset and progression e. g., by clearing amyloid beta (Aβ) through phagocytosis. Genome-wide association studies (GWAS) have revealed that human microglia and CAMs express numerous risk genes for AD, further highlighting their potentially critical role in AD pathogenesis. Microglia and CAMs are tightly controlled by environmental factors, such as the host microbiota. Notably, it was further reported that the composition of the gut microbiota differed between AD patients and healthy individuals. Hence, emerging studies have analyzed the impact of gut bacteria in different preclinical mouse models for AD as well as in clinical studies, potentially enabling promising new therapeutic options.
Collapse
Affiliation(s)
- Philipp Schaible
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Breisacher Str. 64, 79106, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg im Breisgau, Germany
| | - Julia Henschel
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Breisacher Str. 64, 79106, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg im Breisgau, Germany
| | - Daniel Erny
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Breisacher Str. 64, 79106, Freiburg, Germany.
| |
Collapse
|
21
|
Theodorou A, Tsantzali I, Stefanou MI, Sacco S, Katsanos AH, Shoamanesh A, Karapanayiotides T, Koutroulou I, Stamati P, Werring DJ, Cordonnier C, Palaiodimou L, Zompola C, Boviatsis E, Stavrinou L, Frantzeskaki F, Steiner T, Alexandrov AV, Paraskevas GP, Tsivgoulis G. CSF and plasma biomarkers in cerebral amyloid angiopathy: A single-center study and a systematic review/meta-analysis. Eur Stroke J 2025; 10:278-288. [PMID: 38869035 PMCID: PMC11569450 DOI: 10.1177/23969873241260538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 05/14/2024] [Indexed: 06/14/2024] Open
Abstract
INTRODUCTION There are limited data regarding cerebrospinal fluid (CSF) and plasma biomarkers among patients with Cerebral Amyloid Angiopathy (CAA). We sought to investigate the levels of four biomarkers [β-amyloids (Aβ42 and Aβ40), total tau (tau) and phosphorylated tau (p-tau)] in CAA patients compared to healthy controls (HC) and patients with Alzheimer Disease (AD). PATIENTS AND METHODS A systematic review and meta-analysis of published studies, including also a 5 year single-center cohort study, with available data on CSF and plasma biomarkers in symptomatic sporadic CAA versus HC and AD was conducted. Biomarkers' comparisons were investigated using random-effects models based on the ratio of mean (RoM) biomarker concentrations. RoM < 1 and RoM > 1 indicate lower and higher biomarker concentration in CAA compared to another population, respectively. RESULTS We identified nine cohorts, comprising 327 CAA patients (mean age: 71 ± 5 years; women: 45%) versus 336 HC (mean age: 65 ± 5 years; women: 45%) and 384 AD patients (mean age: 68 ± 3 years; women: 53%) with available data on CSF biomarkers. CSF Aβ42 levels [RoM: 0.47; 95% CI: 0.36-0.62; p < 0.0001], Aβ40 levels [RoM: 0.70; 95% CI: 0.63-0.79; p < 0.0001] and the ratio Aβ42/Aβ40 [RoM: 0.62; 95% CI: 0.39-0.98; p = 0.0438] differentiated CAA from HC. CSF Aβ40 levels [RoM: 0.73; 95% CI: 0.64-0.83; p = 0.0003] differentiated CAA from AD. CSF tau and p-tau levels differentiated CAA from HC [RoM: 1.71; 95% CI: 1.41-2.09; p = 0.0002 and RoM: 1.44; 95% CI: 1.20-1.73; p = 0.0014, respectively] and from AD [RoM: 0.65; 95% CI: 0.58-0.72; p < 0.0001 and RoM: 0.64; 95% CI: 0.57-0.71; p < 0.0001, respectively]. Plasma Aβ42 [RoM: 1.14; 95% CI: 0.89-1.45; p = 0.2079] and Aβ40 [RoM: 1.07; 95% CI: 0.91-1.25; p = 0.3306] levels were comparable between CAA and HC. CONCLUSIONS CAA is characterized by a distinct CSF biomarker pattern compared to HC and AD. CSF Aβ40 levels are lower in CAA compared to HC and AD, while tau and p-tau levels are higher in CAA compared to HC, but lower in comparison to AD patients.
Collapse
Affiliation(s)
- Aikaterini Theodorou
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioanna Tsantzali
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria-Ioanna Stefanou
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Simona Sacco
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, Via Vetoio, Italy
| | - Aristeidis H Katsanos
- Division of Neurology, McMaster University/Population Health Research Institute, Hamilton, Canada
| | - Ashkan Shoamanesh
- Division of Neurology, McMaster University/Population Health Research Institute, Hamilton, Canada
| | - Theodoros Karapanayiotides
- Second Department of Neurology, Aristotle University of Thessaloniki, School of Medicine, AHEPA University Hospital, Thessaloniki, Greece
| | - Ioanna Koutroulou
- Second Department of Neurology, Aristotle University of Thessaloniki, School of Medicine, AHEPA University Hospital, Thessaloniki, Greece
| | - Polyxeni Stamati
- Department of Neurology, Laboratory of Neurogenetics, University of Thessaly, University Hospital of Larissa, Biopolis, Mezourlo Hill, Larissa, Greece
| | - David J Werring
- Stroke Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Charlotte Cordonnier
- University Lille, Inserm, CHU Lille, U1172, LilNCog, Lille Neuroscience and Cognition, France
| | - Lina Palaiodimou
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Zompola
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Efstathios Boviatsis
- Second Department of Neurosurgery, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Lampis Stavrinou
- Second Department of Neurosurgery, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Frantzeska Frantzeskaki
- Second Critical Care Department, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Thorsten Steiner
- Departments of Neurology, Klinikum Frankfurt Höchst, Frankfurt and Heidelberg University Hospital, Heidelberg, Germany
| | - Andrei V Alexandrov
- Department of Neurology, University of Arizona, Banner University Medical Center, Phoenix
| | - Georgios P Paraskevas
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgios Tsivgoulis
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Department of Neurology, University of Tennessee Health Science Center, Memphis
| |
Collapse
|
22
|
Cai Y, Kanyo J, Wilson R, Bathla S, Cardozo PL, Tong L, Qin S, Fuentes LA, Pinheiro-de-Sousa I, Huynh T, Sun L, Mansuri MS, Tian Z, Gan HR, Braker A, Trinh HK, Huttner A, Lam TT, Petsalaki E, Brennand KJ, Nairn AC, Grutzendler J. Subcellular proteomics and iPSC modeling uncover reversible mechanisms of axonal pathology in Alzheimer's disease. NATURE AGING 2025; 5:504-527. [PMID: 40065072 PMCID: PMC11922768 DOI: 10.1038/s43587-025-00823-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 01/29/2025] [Indexed: 03/21/2025]
Abstract
Dystrophic neurites (also termed axonal spheroids) are found around amyloid deposits in Alzheimer's disease (AD), where they impair axonal electrical conduction, disrupt neural circuits and correlate with AD severity. Despite their importance, the mechanisms underlying spheroid formation remain incompletely understood. To address this, we developed a proximity labeling approach to uncover the proteome of spheroids in human postmortem and mouse brains. Additionally, we established a human induced pluripotent stem cell (iPSC)-derived AD model enabling mechanistic investigation and optical electrophysiology. These complementary approaches revealed the subcellular molecular architecture of spheroids and identified abnormalities in key biological processes, including protein turnover, cytoskeleton dynamics and lipid transport. Notably, the PI3K/AKT/mTOR pathway, which regulates these processes, was activated in spheroids. Furthermore, phosphorylated mTOR levels in spheroids correlated with AD severity in humans. Notably, mTOR inhibition in iPSC-derived neurons and mice ameliorated spheroid pathology. Altogether, our study provides a multidisciplinary toolkit for investigating mechanisms and therapeutic targets for axonal pathology in neurodegeneration.
Collapse
Affiliation(s)
- Yifei Cai
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.
| | - Jean Kanyo
- Keck MS & Proteomics Resource, Yale School of Medicine, New Haven, CT, USA
| | - Rashaun Wilson
- Keck MS & Proteomics Resource, Yale School of Medicine, New Haven, CT, USA
| | - Shveta Bathla
- Yale/NIDA Neuroproteomics Center, Yale University, New Haven, CT, USA
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | | | - Lei Tong
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Shanshan Qin
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Lukas A Fuentes
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
| | | | - Tram Huynh
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Liyuan Sun
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Mohammad Shahid Mansuri
- Yale/NIDA Neuroproteomics Center, Yale University, New Haven, CT, USA
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Zichen Tian
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Hao-Ran Gan
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Amber Braker
- Yale College, Department of Neuroscience, Yale University, New Haven, CT, USA
| | - Hoang Kim Trinh
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Anita Huttner
- Department of Pathology, Yale University, New Haven, CT, USA
| | - TuKiet T Lam
- Keck MS & Proteomics Resource, Yale School of Medicine, New Haven, CT, USA
- Yale/NIDA Neuroproteomics Center, Yale University, New Haven, CT, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Evangelia Petsalaki
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK
| | - Kristen J Brennand
- Department of Psychiatry, Yale University, New Haven, CT, USA
- Department of Genetics, Yale University, New Haven, CT, USA
| | - Angus C Nairn
- Yale/NIDA Neuroproteomics Center, Yale University, New Haven, CT, USA
- Department of Psychiatry, Yale University, New Haven, CT, USA
- Department of Pharmacology, Yale University, New Haven, CT, USA
| | - Jaime Grutzendler
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.
- Department of Neuroscience, Yale University, New Haven, CT, USA.
- Wu Tsai Institute, Yale University, New Haven, CT, USA.
| |
Collapse
|
23
|
Wang Q, Liu Z, Wang Y, Liu Y, Chen Y, Zhang S, Zeng W, Li D, Yang F, He Z, Xiao W, Liu C, Wang C. Quantitative chemoproteomics reveals dopamine's protective modification of Tau. Nat Chem Biol 2025:10.1038/s41589-025-01849-9. [PMID: 39979588 DOI: 10.1038/s41589-025-01849-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 01/24/2025] [Indexed: 02/22/2025]
Abstract
Dopamine (DA) is one of the most important neurotransmitters. Its oxidation leads to electrophilic quinone, which covalently modifies nucleophilic residues in proteins, resulting in 'dopamination'. Individual dopaminated proteins have been studied, most of which were functionally damaged by dopamination. Here, we developed a quantitative chemoproteomic strategy to site-specifically measure proteins' dopamination. More than 6,000 dopamination sites were quantified. Half-maximal inhibitory concentration values for 63 hypersensitive sites were measured. Among them, hypersensitive dopamination of two cysteines in microtubule-associated protein Tau was biochemically validated and functionally characterized to prevent Tau's amyloid fibrillation and promote Tau-mediated assembly of microtubules. In addition, endogenous dopamination of Tau in mouse brain was detected through targeted mass spectrometry analysis. Our study not only provides a global portrait of dopamination but also discovers a protective role of DA in regulating the function of Tau, which will enhance our understanding of the physiological and pathological functions of DA in human brain.
Collapse
Affiliation(s)
- Qianwen Wang
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Zhengtao Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Youjia Wang
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Yuan Liu
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Ying Chen
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Shengnan Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Wen Zeng
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Dan Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Fan Yang
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Zhuohao He
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Weidi Xiao
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing, China.
- Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, China.
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
- University of the Chinese Academy of Sciences, Beijing, China.
| | - Chu Wang
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing, China.
- Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, China.
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
| |
Collapse
|
24
|
Fayed EA, El-Sebaey SA, Ebrahim MA, Abu-Elfotuh K, El-Sayed Mansour R, Mohamed EK, Hamdan AME, Al-Subaie FT, Albalawi GS, Albalawi TM, Hamdan AM, Mohammed AA, Ramsis TM. Discovery of novel bicyclic and tricyclic cyclohepta[b]thiophene derivatives as multipotent AChE and BChE inhibitors, in-Vivo and in-Vitro assays, ADMET and molecular docking simulation. Eur J Med Chem 2025; 284:117201. [PMID: 39731791 DOI: 10.1016/j.ejmech.2024.117201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/21/2024] [Accepted: 12/21/2024] [Indexed: 12/30/2024]
Abstract
Alzheimer's disease (AD) is primarily caused by oxidative stress, hyperphosphorylated τ-protein aggregation, and amyloid-β deposition. Changes in dopaminergic and serotoninergic neurotransmitter pathways are linked to certain symptoms of AD. Derivatives of bicyclic and tricyclic cyclohepta[b]thiophene were developed to identify new potential candidates as acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) inhibitors for the treatment of AD. All synthesized compounds exhibited AChE inhibition with IC50 values below 15 μM, while all compounds exhibited BChE inhibition with IC50 values below 25 μM. Compounds 9 and 12 exhibited AChE inhibitory activities with IC50 values of 0.51 μM and 0.55 μM, respectively. Compounds 5 and 9 demonstrated excellent inhibitory activity against BChE with IC50 values of 2.9 μM and 2.48 μM, respectively. Compounds 9, 13, and 14 were found to be the most active in terms of the decrease in the escape latency time, with values comparable to that of Donepezil. Compounds 10, 11, and 12 exhibited promising effects on learning and memory. Compounds 5, 10, 11, and 12 exhibited promising SAP values of 70.67 %, 71.5 %, 74.33 % and 73.83 %, respectively. Other biomarkers were evaluated in rat brains including TAC, MDA, SOD, BDNF, IL-β and TNF-α. Fundamental features of ADMET have been computed in-silico for synthesized compounds. Molecular docking was performed to confirm the binding of the novel compounds to the targets.
Collapse
Affiliation(s)
- Eman A Fayed
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, 11754, Egypt.
| | - Samiha Ahmed El-Sebaey
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, 11754, Egypt
| | - Maha A Ebrahim
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, 11754, Egypt
| | - Karema Abu-Elfotuh
- Department of Clinical Pharmacy, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, 11754, Egypt; College of Pharmacy, Al-Ayen Iraqi University, Thi-Qar, 64001, Iraq
| | - Reda El-Sayed Mansour
- Department of Pharmaceutical Medicinal Chemistry and Drug Design, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, 11754, Egypt
| | - Ehsan Khedre Mohamed
- Department of Biochemistry, Egyptian DRUG AUTHORITY (EDA), Formerly National Organization of Drug Control and Research (NODCAR), Giza, Egypt
| | - Ahmed M E Hamdan
- Department of Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, Tabuk, 71491, Saudi Arabia; Prince Fahad bin Sultan Chair for Biomedical Research, University of Tabuk, Saudi Arabia
| | | | | | | | - Amira M Hamdan
- Oceanography Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Asmaa A Mohammed
- Department Pharmacology and Toxicology, Faculty of Pharmacy Girls Al-Azhar University, Cairo, 11754, Egypt
| | - Triveena M Ramsis
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sinai University, Kantara Branch, Ismailia, 41636, Egypt
| |
Collapse
|
25
|
Paul PS, Rathnam M, Khalili A, Cortez LM, Srinivasan M, Planel E, Cho JY, Wille H, Sim VL, Mok SA, Kar S. Temperature-Dependent Aggregation of Tau Protein Is Attenuated by Native PLGA Nanoparticles Under in vitro Conditions. Int J Nanomedicine 2025; 20:1999-2019. [PMID: 39968061 PMCID: PMC11834738 DOI: 10.2147/ijn.s494104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 02/01/2025] [Indexed: 02/20/2025] Open
Abstract
Introduction Hyperphosphorylation and aggregation of the microtubule-associated tau protein, which plays a critical role in many neurodegenerative diseases (ie, tauopathies) including Alzheimer's disease (AD), are known to be regulated by a variety of environmental factors including temperature. In this study we evaluated the effects of FDA-approved poly (D,L-lactide-co-glycolic) acid (PLGA) nanoparticles, which can inhibit amyloid-β aggregation/toxicity in cellular/animal models of AD, on temperature-dependent aggregation of 0N4R tau isoforms in vitro. Methods We have used a variety of biophysical (Thioflavin T kinetics, dynamic light scattering and asymmetric-flow field-flow fractionation), structural (fluorescence imaging and transmission electron microscopy) and biochemical (Filter-trap assay and detection of soluble protein) approaches, to evaluate the effects of native PLGA nanoparticles on the temperature-dependent tau aggregation. Results Our results show that the aggregation propensity of 0N4R tau increases significantly in a dose-dependent manner with a rise in temperature from 27°C to 40°C, as measured by lag time and aggregation rate. Additionally, the aggregation of 2N4R tau increases in a dose-dependent manner. Native PLGA significantly inhibits tau aggregation at all temperatures in a concentration-dependent manner, possibly by interacting with the aggregation-prone hydrophobic hexapeptide motifs of tau. Additionally, native PLGA is able to trigger disassembly of preformed 0N4R tau aggregates as a function of temperature from 27°C to 40°C. Conclusion These results, taken together, suggest that native PLGA nanoparticles can not only attenuate temperature-dependent tau aggregation but also promote disassembly of preformed aggregates, which increased with a rise of temperature. Given the evidence that temperature can influence tau pathology, we believe that native PLGA may have a unique potential to regulate tau abnormalities associated with AD-related pathology.
Collapse
Affiliation(s)
- Pallabi Sil Paul
- Department of Medicine (Neurology), Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, T6G 2M8, Canada
| | - Mallesh Rathnam
- Department of Medicine (Neurology), Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, T6G 2M8, Canada
| | - Aria Khalili
- Quantum and Nanotechnology Research Centre, National Research Council Canada, Edmonton, Alberta, Canada
| | - Leonardo M Cortez
- Department of Medicine (Neurology), Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, T6G 2M8, Canada
| | - Mahalashmi Srinivasan
- Department of Biochemistry, Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada
| | - Emmanuel Planel
- Department of Psychiatry and Neurosciences, University of Laval, Quebec, Canada
| | - Jae-Young Cho
- Quantum and Nanotechnology Research Centre, National Research Council Canada, Edmonton, Alberta, Canada
- Department of Mechanical Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Holger Wille
- Department of Biochemistry, Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada
| | - Valerie L Sim
- Department of Medicine (Neurology), Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, T6G 2M8, Canada
| | - Sue-Ann Mok
- Department of Biochemistry, Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada
| | - Satyabrata Kar
- Department of Medicine (Neurology), Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, T6G 2M8, Canada
| |
Collapse
|
26
|
Yarbro JM, Han X, Dasgupta A, Yang K, Liu D, Shrestha HK, Zaman M, Wang Z, Yu K, Lee DG, Vanderwall D, Niu M, Sun H, Xie B, Chen PC, Jiao Y, Zhang X, Wu Z, Chepyala SR, Fu Y, Li Y, Yuan ZF, Wang X, Poudel S, Vagnerova B, He Q, Tang A, Ronaldson PT, Chang R, Yu G, Liu Y, Peng J. Human and mouse proteomics reveals the shared pathways in Alzheimer's disease and delayed protein turnover in the amyloidome. Nat Commun 2025; 16:1533. [PMID: 39934151 PMCID: PMC11814087 DOI: 10.1038/s41467-025-56853-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 02/04/2025] [Indexed: 02/13/2025] Open
Abstract
Murine models of Alzheimer's disease (AD) are crucial for elucidating disease mechanisms but have limitations in fully representing AD molecular complexities. Here we present the comprehensive, age-dependent brain proteome and phosphoproteome across multiple mouse models of amyloidosis. We identified shared pathways by integrating with human metadata and prioritized components by multi-omics analysis. Collectively, two commonly used models (5xFAD and APP-KI) replicate 30% of the human protein alterations; additional genetic incorporation of tau and splicing pathologies increases this similarity to 42%. We dissected the proteome-transcriptome inconsistency in AD and 5xFAD mouse brains, revealing that inconsistent proteins are enriched within amyloid plaque microenvironment (amyloidome). Our analysis of the 5xFAD proteome turnover demonstrates that amyloid formation delays the degradation of amyloidome components, including Aβ-binding proteins and autophagy/lysosomal proteins. Our proteomic strategy defines shared AD pathways, identifies potential targets, and underscores that protein turnover contributes to proteome-transcriptome discrepancies during AD progression.
Collapse
Affiliation(s)
- Jay M Yarbro
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xian Han
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Abhijit Dasgupta
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Computer Science and Engineering, SRM University AP, Andhra Pradesh, India
| | - Ka Yang
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Danting Liu
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Him K Shrestha
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Masihuz Zaman
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Zhen Wang
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Kaiwen Yu
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Dong Geun Lee
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - David Vanderwall
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Mingming Niu
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Huan Sun
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Boer Xie
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ping-Chung Chen
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yun Jiao
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xue Zhang
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Zhiping Wu
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Surendhar R Chepyala
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yingxue Fu
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yuxin Li
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Zuo-Fei Yuan
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xusheng Wang
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Suresh Poudel
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Barbora Vagnerova
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Qianying He
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Andrew Tang
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Patrick T Ronaldson
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Rui Chang
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Gang Yu
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yansheng Liu
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
- Yale Cancer Biology Institute, Yale University School of Medicine, West Haven, CT, USA
- Department of Biomedical Informatics & Data Science, Yale University School of Medicine, West Haven, CT, USA
| | - Junmin Peng
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA.
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, USA.
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
27
|
Calvin-Dunn KN, Mcneela A, Leisgang Osse A, Bhasin G, Ridenour M, Kinney JW, Hyman JM. Electrophysiological insights into Alzheimer's disease: A review of human and animal studies. Neurosci Biobehav Rev 2025; 169:105987. [PMID: 39732222 DOI: 10.1016/j.neubiorev.2024.105987] [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: 01/22/2024] [Revised: 11/16/2024] [Accepted: 12/17/2024] [Indexed: 12/30/2024]
Abstract
This review highlights the crucial role of neuroelectrophysiology in illuminating the mechanisms underlying Alzheimer's disease (AD) pathogenesis and progression, emphasizing its potential to inform the development of effective treatments. Electrophysiological techniques provide unparalleled precision in exploring the intricate networks affected by AD, offering insights into the synaptic dysfunction, network alterations, and oscillatory abnormalities that characterize the disease. We discuss a range of electrophysiological methods, from non-invasive clinical techniques like electroencephalography and magnetoencephalography to invasive recordings in animal models. By drawing on findings from these studies, we demonstrate how electrophysiological research has deepened our understanding of AD-related network disruptions, paving the way for targeted therapeutic interventions. Moreover, we underscore the potential of electrophysiological modalities to play a pivotal role in evaluating treatment efficacy. Integrating electrophysiological data with clinical neuroimaging and longitudinal studies holds promise for a more comprehensive understanding of AD, enabling early detection and the development of personalized treatment strategies. This expanded research landscape offers new avenues for unraveling the complexities of AD and advancing therapeutic approaches.
Collapse
Affiliation(s)
- Kirsten N Calvin-Dunn
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States; Cleveland Clinic Lou Ruvo Center for Brain Health, United States.
| | - Adam Mcneela
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States
| | - A Leisgang Osse
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States; Department of Brain Health, University of Nevada, Las Vegas, United States
| | - G Bhasin
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States; Department of Psychology, University of Nevada, Las Vegas, United States
| | - M Ridenour
- Department of Psychology, University of Nevada, Las Vegas, United States
| | - J W Kinney
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States; Department of Brain Health, University of Nevada, Las Vegas, United States
| | - J M Hyman
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas, United States; Department of Psychology, University of Nevada, Las Vegas, United States
| |
Collapse
|
28
|
Shevchuk DV, Tukhvatulin AI, Dzharullaeva AS, Berdalina IA, Zakharova MN. Molecular Biomarkers of Neurodegeneration in Amyotrophic Lateral Sclerosis. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:276-288. [PMID: 40254405 DOI: 10.1134/s0006297924604039] [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: 11/29/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 04/22/2025]
Abstract
Amyotrophic lateral sclerosis (ALS) is the most prevalent motor neuron disease. However, definitive diagnosis could be delayed by up to 12 months due to the lack of specific and sensitive biomarkers for ALS. In our study, conducted for the first time on a large cohort of ALS patients (n = 100) within the Russian population, we assessed key biomarkers of neurodegenerative pathology, including β-amyloids (Aβ40 and Aβ42) and tau proteins (Tau-total and Tau-p181), as well as other pathogenetically relevant, promising biomarkers such as FGF-21, Kallikrein-6 (KLK-6), NCAM-1, Neurogranin (NRGN), TDP-43, Apolipoprotein E4, Clusterin (Apo J), Complement Factor H, Fetuin-A, α2-Macroglobulin, Apo AI, Apo CIII, Apo E, Complement C3, GDNF, sRAGE, and S100B protein. Significant differences between the ALS patients and the control group were observed for Aβ40 (p = 0.044), Aβ42 (p < 0.001), FGF-21 (p < 0.001), Tau-total (p = 0.001), Tau-p181 (p = 0.014), Clusterin (p < 0.001), Complement C3 (p = 0.001), and S100B (p = 0.024). A significant direct correlation was found between the ALSFRS-R score and concentrations of Aβ40 and Aβ42. Changes in the complement system (Complement C3 and Complement Factor H) were identified, highlighting critical role of neuroinflammatory processes in ALS pathogenesis. Additionally, increased levels of FGF-21 were observed in the patients with the bulbar onset of ALS. Significant increase in the concentration of the chaperone protein clusterin in the patients with rapid disease progression suggests its potential as a prognostic biomarker for motor neuron disease. Furthermore, its role in maintaining proteostasis could provide novel therapeutic targets.
Collapse
Affiliation(s)
| | - Amir I Tukhvatulin
- Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, 123098, Russia
| | - Alina S Dzharullaeva
- Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, 123098, Russia
| | | | | |
Collapse
|
29
|
Tao Y, Xu P, Zhang S, Shangguan W, Yang G, Liu K, Li X, Sun Y, Zhao Q, Li D, Yu B, Liu C. Time-course remodeling and pathology intervention of α-synuclein amyloid fibril by heparin and heparin-like oligosaccharides. Nat Struct Mol Biol 2025; 32:369-380. [PMID: 39420234 DOI: 10.1038/s41594-024-01407-2] [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: 02/16/2023] [Accepted: 09/25/2024] [Indexed: 10/19/2024]
Abstract
Amyloid fibrils represent a pathological state of protein polymer that is closely associated with various neurodegenerative diseases. Polysaccharides have a prominent role in recognizing amyloid fibrils and mediating their pathogenicity. However, the mechanism underlying the amyloid-polysaccharide interaction remains elusive. We also do not know its impact on the structure and pathology of formed fibrils. Here, we used cryo-electron microscopy to analyze the atomic structures of mature α-synuclein (α-syn) fibrils upon binding with polymeric heparin and heparin-like oligosaccharides. The fibril structure, including the helical twist and conformation of α-syn, changed over time upon the binding of heparin but not oligosaccharides. The sulfation pattern and numbers of saccharide units are important for the binding. Similarly, negatively charged biopolymers typically interact with amyloid fibrils, including tau and various α-syn polymorphs, leading to alterations in their conformation. Moreover, we show that heparin-like oligosaccharides can not only block neuronal uptake and propagation of formed α-syn fibrils but also inhibit α-syn fibrillation. This work demonstrates a distinctive activity of heparin and biopolymers in remodeling amyloid fibrils and suggests the pharmaceutical potential of heparin-like oligosaccharides.
Collapse
Affiliation(s)
- Youqi Tao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Peng Xu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shenqing Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Shangguan
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Guang Yang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Kaien Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Xiang Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai, China
| | - Yunpeng Sun
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Qinyue Zhao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai, China
| | - Dan Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai, China
| | - Biao Yu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Cong Liu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
30
|
Abdelhamid M, Counts SE, Zhou C, Hida H, Kim JI, Michikawa M, Jung CG. Protective Effects of Bifidobacterium Breve MCC1274 as a Novel Therapy for Alzheimer's Disease. Nutrients 2025; 17:558. [PMID: 39940416 PMCID: PMC11820889 DOI: 10.3390/nu17030558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 01/14/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and is characterized by memory impairment that significantly interferes with daily life. Therapeutic options for AD that substantively modify disease progression remain a critical unmet need. In this regard, the gut microbiota is crucial in maintaining human health by regulating metabolism and immune responses, and increasing evidence suggests that probiotics, particularly beneficial bacteria, can enhance memory and cognitive functions. Recent studies have highlighted the positive effects of Bifidobacterium breve MCC1274 (B. breve MCC1274) on individuals with mild cognitive impairment (MCI) and schizophrenia. Additionally, oral supplementation with B. breve MCC1274 has been shown to effectively prevent memory decline in AppNL-G-F mice. In relation to Alzheimer's pathology, oral supplementation with B. breve MCC1274 has been found to reduce amyloid-β (Aβ) accumulation and tau phosphorylation in both AppNL-G-F and wild-type (WT) mice. It also decreases microglial activation and increases levels of synaptic proteins. In this review, we examine the beneficial effects of B. breve MCC1274 on AD, exploring potential mechanisms of action and how this probiotic strain may aid in preventing or treating the disease. Furthermore, we discuss the broader implications of B. breve MCC1274 for improving overall host health and provide insights into future research directions for this promising probiotic therapy.
Collapse
Affiliation(s)
- Mona Abdelhamid
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Avenue NW, Grand Rapids, MI 49503, USA; (M.A.); (S.E.C.)
| | - Scott E. Counts
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Avenue NW, Grand Rapids, MI 49503, USA; (M.A.); (S.E.C.)
| | - Chunyu Zhou
- Department of Biochemistry, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan;
| | - Hideki Hida
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan;
| | - Jae-Il Kim
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea;
| | - Makoto Michikawa
- Department of Geriatric Medicine, School of Life Dentistry at Niigata, Nippon Dental University, Niigata 951-8580, Japan
| | - Cha-Gyun Jung
- Center for Nursing International Promotion, Nagoya City University Graduate School of Nursing, Nagoya 467-8601, Japan
| |
Collapse
|
31
|
Hyde VR, Zhou C, Fernandez JR, Chatterjee K, Ramakrishna P, Lin A, Fisher GW, Çeliker OT, Caldwell J, Bender O, Sauer PJ, Lugo-Martinez J, Bar DZ, D'Aiuto L, Shemesh OA. Anti-herpetic tau preserves neurons via the cGAS-STING-TBK1 pathway in Alzheimer's disease. Cell Rep 2025; 44:115109. [PMID: 39753133 DOI: 10.1016/j.celrep.2024.115109] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 08/06/2024] [Accepted: 12/03/2024] [Indexed: 02/01/2025] Open
Abstract
Alzheimer's disease (AD) diagnosis relies on the presence of extracellular β-amyloid (Aβ) and intracellular hyperphosphorylated tau (p-tau). Emerging evidence suggests a potential link between AD pathologies and infectious agents, with herpes simplex virus 1 (HSV-1) being a leading candidate. Our investigation, using metagenomics, mass spectrometry, western blotting, and decrowding expansion pathology, detects HSV-1-associated proteins in human brain samples. Expression of the herpesvirus protein ICP27 increases with AD severity and strongly colocalizes with p-tau but not with Aβ. Modeling in human brain organoids shows that HSV-1 infection elevates tau phosphorylation. Notably, p-tau reduces ICP27 expression and markedly decreases post-infection neuronal death from 64% to 7%. This modeling prompts investigation into the cGAS-STING-TBK1 pathway products, nuclear factor (NF)-κB and IRF-3, which colocalizes with ICP27 and p-tau in AD. Furthermore, experimental activation of STING enhances tau phosphorylation, while TBK1 inhibition prevents it. Together, these findings suggest that tau phosphorylation acts as an innate immune response in AD, driven by cGAS-STING.
Collapse
Affiliation(s)
- Vanesa R Hyde
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Chaoming Zhou
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Juan R Fernandez
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Krishnashis Chatterjee
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Pururav Ramakrishna
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Amanda Lin
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Gregory W Fisher
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Orhan Tunç Çeliker
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jill Caldwell
- Department of Psychiatry, Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Omer Bender
- Department of Oral Biology, Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Peter Joseph Sauer
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Jose Lugo-Martinez
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Daniel Z Bar
- Department of Oral Biology, Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Leonardo D'Aiuto
- Department of Psychiatry, Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Or A Shemesh
- School of Pharmacy, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112102, Israel; Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
| |
Collapse
|
32
|
Chen X, Tian Q, Gao M, Zhou X, Zhou C. Lingguizhugan decoction enhances autophagy of Alzheimer's disease via regulating the mTOR/ p70s6K pathway in vivo and in vitro. Front Aging Neurosci 2025; 17:1478199. [PMID: 39911633 PMCID: PMC11794305 DOI: 10.3389/fnagi.2025.1478199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 01/08/2025] [Indexed: 02/07/2025] Open
Abstract
Introduction Lingguizhugan decoction (LGZG) has been reported to treat Alzheimer's disease (AD) by anti-inflammatory and transporting amyloid-β (Aβ). Methods Using APP/PS1 transgenic mice as in vivo model and gave LGZG decoction by oral gavage. Using Aβ25-35-induced SH-SY5Y cells as in vitro model and then added LGZG medicated serum (LMS) to observe the regulatory effect of LGZG on AD autophagy-related pathways. Morris water maze (MWM) was used to evaluate the mice's learning and memory ability. Mice's hippocampus tissue sections were stained immunohistochemically to observe hippocampal Aβ deposition. Transmission electron microscopy monitored autophagosomes and autolysosomes. Western blot analysis measured protein expression levels of beclin-1, p62 and light chain 3II (LC3 II) and mTOR signaling. Results: LGZG could greatly improve learning and memory ability of APP/PS1 mice, and enhance autophagy in vitro and in vivo. LGZG increased the levels of beclin-1 and LC3 II and decreased the levels of p62. Conclusion LGZG enhanced autophagy and showed therapeutic potential in AD by inhibiting mTOR/p70s6K signaling.
Collapse
Affiliation(s)
| | | | | | - Xibin Zhou
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chunxiang Zhou
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
33
|
Huang S, Nunez J, Toresco DL, Wen C, Slotabec L, Wang H, Zhang H, Rouhi N, Adenawoola MI, Li J. Alterations in the inflammatory homeostasis of aging-related cardiac dysfunction and Alzheimer's diseases. FASEB J 2025; 39:e70303. [PMID: 39758048 DOI: 10.1096/fj.202402725rr] [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: 11/05/2024] [Revised: 12/20/2024] [Accepted: 12/26/2024] [Indexed: 01/07/2025]
Abstract
Alzheimer's disease (AD) is well known among the elderly and has a profound impact on both patients and their families. Increasing research indicates that AD is a systemic disease, with a strong connection to cardiovascular disease. They share common genetic factors, such as mutations in the presenilin (PS1 and PS2) and the apolipoprotein E (APOE) genes. Cardiovascular conditions can lead to reduced cerebral blood flow and increased oxidative stress. These factors contribute to the accumulation of Aβ plaques and the formation of abnormal tau protein tangles, which are both key pathological features of AD. Additionally, Aβ deposits and abnormal protein responses have been observed in cardiomyocytes as well as in peripheral tissues. The toxic Aβ deposition intensifies damage to the microvascular structure associated with blood-brain barrier disruption and the initiation of neuroinflammation, which may accelerate the onset of neurocognitive deficits and cardiovascular dysfunction. Thus, we discuss the main mechanisms linking AD and cardiac dysfunction to enhance our understanding of these conditions. Ultimately, insights into the brain-heart axis may help us develop effective treatment strategies in the future.
Collapse
Affiliation(s)
- Shuli Huang
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Jeremiah Nunez
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi, USA
| | - Dai Lan Toresco
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi, USA
| | - Changhong Wen
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi, USA
| | - Lily Slotabec
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi, USA
| | - Hao Wang
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Haibei Zhang
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Nadiyeh Rouhi
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Michael I Adenawoola
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Ji Li
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
34
|
Hu Z, Yang J, Zhang S, Li M, Zuo C, Mao C, Zhang Z, Tang M, Shi C, Xu Y. AAV mediated carboxyl terminus of Hsp70 interacting protein overexpression mitigates the cognitive and pathological phenotypes of APP/PS1 mice. Neural Regen Res 2025; 20:253-264. [PMID: 38767490 PMCID: PMC11246129 DOI: 10.4103/nrr.nrr-d-23-01277] [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: 07/31/2023] [Revised: 11/27/2023] [Accepted: 01/02/2024] [Indexed: 05/22/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202501000-00033/figure1/v/2024-05-14T021156Z/r/image-tiff The E3 ubiquitin ligase, carboxyl terminus of heat shock protein 70 (Hsp70) interacting protein (CHIP), also functions as a co-chaperone and plays a crucial role in the protein quality control system. In this study, we aimed to investigate the neuroprotective effect of overexpressed CHIP on Alzheimer's disease. We used an adeno-associated virus vector that can cross the blood-brain barrier to mediate CHIP overexpression in APP/PS1 mouse brain. CHIP overexpression significantly ameliorated the performance of APP/PS1 mice in the Morris water maze and nest building tests, reduced amyloid-β plaques, and decreased the expression of both amyloid-β and phosphorylated tau. CHIP also alleviated the concentration of microglia and astrocytes around plaques. In APP/PS1 mice of a younger age, CHIP overexpression promoted an increase in ADAM10 expression and inhibited β-site APP cleaving enzyme 1, insulin degrading enzyme, and neprilysin expression. Levels of HSP70 and HSP40, which have functional relevance to CHIP, were also increased. Single nuclei transcriptome sequencing in the hippocampus of CHIP overexpressed mice showed that the lysosomal pathway and oligodendrocyte-related biological processes were up-regulated, which may also reflect a potential mechanism for the neuroprotective effect of CHIP. Our research shows that CHIP effectively reduces the behavior and pathological manifestations of APP/PS1 mice. Indeed, overexpression of CHIP could be a beneficial approach for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Zhengwei Hu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jing Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
- NHC Key Laboratory of Prevention and treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Shuo Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Mengjie Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Chunyan Zuo
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Chengyuan Mao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
- NHC Key Laboratory of Prevention and treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Zhongxian Zhang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Mibo Tang
- Department of Gerontology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Changhe Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
- NHC Key Laboratory of Prevention and treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
- NHC Key Laboratory of Prevention and treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan Province, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
35
|
Gheni G, Shinohara M, Masuda‐Suzukake M, Shindo A, Watanabe A, Kawai K, Bu G, Tomimoto H, Hasegawa M, Sato N. Cerebral hypoperfusion reduces tau accumulation. Ann Clin Transl Neurol 2025; 12:69-85. [PMID: 39621511 PMCID: PMC11752094 DOI: 10.1002/acn3.52247] [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: 06/25/2024] [Revised: 10/03/2024] [Accepted: 10/24/2024] [Indexed: 01/22/2025] Open
Abstract
OBJECTIVE Alzheimer's disease (AD) often coexists with cerebrovascular diseases. However, the impact of cerebrovascular diseases such as stroke on AD pathology remains poorly understood. METHODS This study examines the correlation between cerebrovascular diseases and AD pathology. The research was carried out using clinical and neuropathological data collected from the National Alzheimer's Coordinating Center (NACC) database and an animal model in which bilateral common carotid artery stenosis surgery was performed, following the injection of tau seeds into the brains of wild-type mice. RESULTS Analysis of the NACC database suggests that clinical stroke history and lacunar infarcts are associated with lower neurofibrillary tangle pathology. An animal model demonstrates that chronic cerebral hypoperfusion reduces tau pathology, which was observed in not only neurons but also astrocytes, microglia, and oligodendrocytes. Furthermore, we found that astrocytes and microglia were activated in response to tau pathology and chronic cerebral hypoperfusion. Additionally, cerebral hypoperfusion increased a lysosomal enzyme, cathepsin D. INTERPRETATION These data together indicate that cerebral hypoperfusion reduces tau accumulation likely through an increase in microglial phagocytic activity towards tau and an elevation in degradation through cathepsin D. This study contributes to understanding the relationship between tau pathology and cerebrovascular diseases in older people with multimorbidity.
Collapse
Grants
- MEXT15K15272 Grants-in-Aid from Japan Promotion of Science; the Japanese Ministry of Education, Culture, Sports, Science, and Technology
- 19-9 Funding for Longevity Sciences from the National Center for Geriatrics and Gerontology
- P20 AG068053 NIA NIH HHS
- P30 AG062421 NIA NIH HHS
- Takeda Medical Research Foundation Research
- P30 AG066508 NIA NIH HHS
- P30 AG072973 NIA NIH HHS
- P30 AG066530 NIA NIH HHS
- Novartis Foundation for Gerontological Research Award
- P30 AG066509 NIA NIH HHS
- P30 AG066546 NIA NIH HHS
- Mitsui Sumitomo Insurance Welfare Foundation
- P30 AG072979 NIA NIH HHS
- P20 AG068082 NIA NIH HHS
- P30 AG072975 NIA NIH HHS
- P30 AG066444 NIA NIH HHS
- P30 AG066507 NIA NIH HHS
- P30 AG072946 NIA NIH HHS
- P30 AG066518 NIA NIH HHS
- P30 AG066511 NIA NIH HHS
- U24 AG072122 NIA NIH HHS
- MEXT21H02844 Grants-in-Aid from Japan Promotion of Science; the Japanese Ministry of Education, Culture, Sports, Science, and Technology
- P30 AG066512 NIA NIH HHS
- MEXT26293167 Grants-in-Aid from Japan Promotion of Science; the Japanese Ministry of Education, Culture, Sports, Science, and Technology
- P30 AG066515 NIA NIH HHS
- P30 AG072978 NIA NIH HHS
- P30 AG062429 NIA NIH HHS
- P30 AG066519 NIA NIH HHS
- Takeda Science Foundation Research Encouragement Grant
- 28-45 Funding for Longevity Sciences from the National Center for Geriatrics and Gerontology
- P30 AG062422 NIA NIH HHS
- R01 AG079280 NIA NIH HHS
- P30 AG066462 NIA NIH HHS
- 19-3 Funding for Longevity Sciences from the National Center for Geriatrics and Gerontology
- 21-12 Funding for Longevity Sciences from the National Center for Geriatrics and Gerontology
- MEXT24K02361 Grants-in-Aid from Japan Promotion of Science; the Japanese Ministry of Education, Culture, Sports, Science, and Technology
- P20 AG068077 NIA NIH HHS
- P30 AG072977 NIA NIH HHS
- P30 AG062677 NIA NIH HHS
- P20 AG068024 NIA NIH HHS
- P30 AG072958 NIA NIH HHS
- P30 AG062715 NIA NIH HHS
- P30 AG066506 NIA NIH HHS
- P30 AG066468 NIA NIH HHS
- Annual Research Award Grant from the Japanese Society of Anti-aging Medicine
- P30 AG072976 NIA NIH HHS
- P30 AG072947 NIA NIH HHS
- P30 AG072931 NIA NIH HHS
- MEXT17H04154 Grants-in-Aid from Japan Promotion of Science; the Japanese Ministry of Education, Culture, Sports, Science, and Technology
- SENSHIN Medical Research Foundation Research Grant
- P30 AG072972 NIA NIH HHS
- P30 AG066514 NIA NIH HHS
- P30 AG072959 NIA NIH HHS
- 24-16 Funding for Longevity Sciences from the National Center for Geriatrics and Gerontology
- Mitsui Sumitomo Insurance Welfare Foundation
Collapse
Affiliation(s)
- Ghupurjan Gheni
- Department of Aging Neurobiology, Center for Development of Advanced Medicine for DementiaNational Center for Geriatrics and Gerontology7‐430 MoriokaObuAichi474‐8511Japan
| | - Mitsuru Shinohara
- Department of Aging Neurobiology, Center for Development of Advanced Medicine for DementiaNational Center for Geriatrics and Gerontology7‐430 MoriokaObuAichi474‐8511Japan
- Department of NeuroscienceMayo ClinicJacksonvilleFlorida32224USA
| | - Masami Masuda‐Suzukake
- Dementia Research ProjectTokyo Metropolitan Institute of Medical Science2‐1‐6 Kamikitazawa, Setagaya‐kuTokyo156‐8506Japan
| | - Akihiko Shindo
- Department of Neurology, Graduate School of MedicineMie University1577 Kurima MachiyachoTsu CityMie514‐8507Japan
| | - Atsushi Watanabe
- Equipment Management Division, Center for Core Facility Administration, National Center for Geriatrics and Gerontology7‐430 MoriokaObuAichi474‐8511Japan
| | - Kaori Kawai
- Department of Aging Neurobiology, Center for Development of Advanced Medicine for DementiaNational Center for Geriatrics and Gerontology7‐430 MoriokaObuAichi474‐8511Japan
| | - Guojun Bu
- Department of NeuroscienceMayo ClinicJacksonvilleFlorida32224USA
- Division of Life ScienceThe Hong Kong University of Science and TechnologyHong KongChina
| | - Hidekazu Tomimoto
- Department of Neurology, Graduate School of MedicineMie University1577 Kurima MachiyachoTsu CityMie514‐8507Japan
| | - Masato Hasegawa
- Dementia Research ProjectTokyo Metropolitan Institute of Medical Science2‐1‐6 Kamikitazawa, Setagaya‐kuTokyo156‐8506Japan
| | - Naoyuki Sato
- Department of Aging Neurobiology, Center for Development of Advanced Medicine for DementiaNational Center for Geriatrics and Gerontology7‐430 MoriokaObuAichi474‐8511Japan
| |
Collapse
|
36
|
Park HH, Kim BH, Leem SH, Park YH, Chung H, Yoo DH, Park I, Nam Y, Kim S, Shin SJ, Moon M. Therapeutic Potential of Traditional Oriental Medicines in Targeting Tau Pathology: Insights from Cell-free and Cell-based Screening. Curr Med Chem 2025; 32:1830-1845. [PMID: 38486385 DOI: 10.2174/0109298673295901240311072440] [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: 12/29/2023] [Revised: 02/08/2024] [Accepted: 02/27/2024] [Indexed: 05/13/2025]
Abstract
BACKGROUND Traditional Oriental Medicines (TOMs) formulated using a variety of medicinal plants have a low risk of side effects. In previous studies, five TOMs, namely Dangguijakyaksan, Hwanglyeonhaedoktang, Ukgansan, Palmijihwanghwan, and Jowiseungchungtang have been commonly used to treat patients with Alzheimer's disease (AD). However, only a few studies have investigated the effects of these five TOMs on tau pathology. OBJECTIVE This study aimed to examine the effect of five TOMs on various tau pathologies, including post-translational modifications, aggregation and deposition, tau-induced neurotoxicity, and tau-induced neuroinflammation. METHODS Immunocytochemistry was used to investigate the hyperphosphorylation of tau induced by okadaic acid. In addition, the thioflavin T assay was used to assess the effects of the TOMs on the inhibition of tau K18 aggregation and the dissociation of tau K18 aggregates. Moreover, a water-soluble tetrazolium-1 assay and a quantitative reverse transcription polymerase chain reaction were used to evaluate the effects of the TOMs on tau-induced neurotoxicity and inflammatory cytokines in HT22 and BV2 cells, respectively. RESULTS The five TOMs investigated in this study significantly reduced okadaic acid-induced tau hyperphosphorylation. Hwanglyeonhaedoktang inhibited the aggregation of tau and promoted the dissociation of tau aggregates. Dangguijakyaksan and Hwanglyeonhaedoktang attenuated tau-induced neurotoxicity in HT22 cells. In addition, Dangguijakyaksan, Hwanglyeonhaedoktang, Ukgansan, and Palmijihwanghwan reduced tauinduced pro-inflammatory cytokine levels in BV2 cells. CONCLUSION Our results suggest that five TOMs are potential therapeutic candidates for tau pathology. In particular, Hwanglyeonhaedoktang showed the greatest efficacy among the five TOMs in cell-free and cell-based screening approaches. These findings suggest that Hwanglyeonhaedoktang is suitable for treating AD patients with tau pathology.
Collapse
Grants
- NRF-2018R1D1A3B07041059, RS-2023-00240010, NRF-2022R1A6A3A13053190, RS-2023-00212388, RS-2023-00273557, NRF-2022R1I1A3052949 Basic Science Research Program of the National Research Foundation of Korea (NRF), by the Ministry of Science, ICT & Future Planning
- HF21C0021, HI23C1263 Korea Health Technology R&D Project through, Korea Health Industry Development Institute (KHIDI), by the Ministry of Health & Welfare, Republic of Korea
Collapse
Affiliation(s)
- Hyun Ha Park
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
| | - Byeong-Hyeon Kim
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
| | - Seol Hwa Leem
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
| | - Yong Ho Park
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
| | - Hyunju Chung
- Department of Core Research Laboratory, Medical Science Research Institute, Kyung Hee University Hospital Gangdong, Seoul, 05278, Republic of Korea
| | - Doo-Han Yoo
- Research Institute for Dementia Science, Konyang University, 158, Gwanjeodong-ro Seo-gu, Daejeon, 35365, Republic of Korea
- Department of Occupational Therapy, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
| | - Insu Park
- Department of Biomedical Engineering, Konyang University, 158, Gwanjeodong- ro, Seo-gu, Daejeon, 35365, Republic of Korea
| | - Yunkwon Nam
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
| | - Sujin Kim
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
- Research Institute for Dementia Science, Konyang University, 158, Gwanjeodong-ro Seo-gu, Daejeon, 35365, Republic of Korea
| | - Soo Jung Shin
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
- Research Institute for Dementia Science, Konyang University, 158, Gwanjeodong-ro Seo-gu, Daejeon, 35365, Republic of Korea
| |
Collapse
|
37
|
Amabebe E, Huang Z, Jash S, Krishnan B, Cheng S, Nakashima A, Li Y, Li Z, Wang R, Menon R, Zhou XZ, Lu KP, Sharma S. Novel Role of Pin1-Cis P-Tau-ApoE Axis in the Pathogenesis of Preeclampsia and Its Connection with Dementia. Biomedicines 2024; 13:29. [PMID: 39857613 PMCID: PMC11763151 DOI: 10.3390/biomedicines13010029] [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: 10/23/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
Preeclampsia (preE) is a severe multisystem hypertensive syndrome of pregnancy associated with ischemia/hypoxia, angiogenic imbalance, apolipoprotein E (ApoE)-mediated dyslipidemia, placental insufficiency, and inflammation at the maternal-fetal interface. Our recent data further suggest that preE is associated with impaired autophagy, vascular dysfunction, and proteinopathy/tauopathy disorder, similar to neurodegenerative diseases such as Alzheimer's disease (AD), including the presence of the cis stereo-isoform of phosphorylated tau (cis P-tau), amyloid-β, and transthyretin in the placenta and circulation. This review provides an overview of the factors that may lead to the induction and accumulation of cis P-tau-like proteins by focusing on the inactivation of peptidyl-prolyl cis-trans isomerase (Pin1) that catalyzes the cis to trans isomerization of P-tau. We also highlighted the novel role of the Pin1-cis P-tau-ApoE axis in the development of preE, and propagation of cis P-tau-mediated abnormal protein aggregation (tauopathy) from the placenta to cerebral tissues later in life, leading to neurodegenerative conditions. In the case of preE, proteinopathy/tauopathy may interrupt trophoblast differentiation and induce cell death, similar to the events occurring in neurons. These events may eventually damage the endothelium and cause systemic features of disorders such as preE. Despite impressive research and therapeutic advances in both fields of preE and neurodegenerative diseases, further investigation of Pin1-cis P-tau and ApoE-related mechanistic underpinnings may unravel novel therapeutic options, and new transcriptional and proteomic markers. This review will also cover genetic polymorphisms in the ApoE alleles leading to dyslipidemia induction that may regulate the pathways causing preE or dementia-like features in the reproductive age or later in life, respectively.
Collapse
Affiliation(s)
- Emmanuel Amabebe
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; (E.A.); (Z.H.); (R.M.)
| | - Zheping Huang
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; (E.A.); (Z.H.); (R.M.)
| | - Sukanta Jash
- Department of Molecular Biology, Cell Biology and Biochemistry, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA;
| | - Balaji Krishnan
- Mitchell Center for Neurodegenerative Diseases, Department of Neurology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA;
| | - Shibin Cheng
- Department of Pediatrics, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA;
| | - Akitoshi Nakashima
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Toyama, Toyama 930-8555, Japan;
| | - Yitong Li
- Departments of Biochemistry and Oncology, Schulich School of Medicine and Dentistry, Robarts Research Institute, Western University, London, ON N6A 3K7, Canada; (Y.L.); (Z.L.); (R.W.); (X.Z.Z.); (K.P.L.)
| | - Zhixong Li
- Departments of Biochemistry and Oncology, Schulich School of Medicine and Dentistry, Robarts Research Institute, Western University, London, ON N6A 3K7, Canada; (Y.L.); (Z.L.); (R.W.); (X.Z.Z.); (K.P.L.)
| | - Ruizhi Wang
- Departments of Biochemistry and Oncology, Schulich School of Medicine and Dentistry, Robarts Research Institute, Western University, London, ON N6A 3K7, Canada; (Y.L.); (Z.L.); (R.W.); (X.Z.Z.); (K.P.L.)
| | - Ramkumar Menon
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; (E.A.); (Z.H.); (R.M.)
| | - Xiao Zhen Zhou
- Departments of Biochemistry and Oncology, Schulich School of Medicine and Dentistry, Robarts Research Institute, Western University, London, ON N6A 3K7, Canada; (Y.L.); (Z.L.); (R.W.); (X.Z.Z.); (K.P.L.)
- Departments of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, Lawson Health Research Institute, Western University, London, ON N6A 3K7, Canada
| | - Kun Ping Lu
- Departments of Biochemistry and Oncology, Schulich School of Medicine and Dentistry, Robarts Research Institute, Western University, London, ON N6A 3K7, Canada; (Y.L.); (Z.L.); (R.W.); (X.Z.Z.); (K.P.L.)
| | - Surendra Sharma
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; (E.A.); (Z.H.); (R.M.)
| |
Collapse
|
38
|
Vermeulen S, Ernst S, Blondeel E, Xia Z, Rappu P, Heino J, Dedeyne S, Denys H, Sys G, Gijsels S, Depypere H, Tummers P, Ceelen W, Craciun L, Demetter P, Raes O, Hendrix A, Van der Eycken J, De Wever O. Pelophen B is a non-taxoid binding microtubule-stabilizing agent with promising preclinical anticancer properties. Sci Rep 2024; 14:30188. [PMID: 39633082 PMCID: PMC11618378 DOI: 10.1038/s41598-024-80672-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024] Open
Abstract
Taxanes, such as paclitaxel (PTX), stabilize microtubules and are used as a first-line therapy in multiple cancer types. Disruption of microtubule equilibrium, which plays an essential role in mitosis and cell homeostasis, ultimately results in cell death. Even though PTX is a very potent chemotherapy, its use is associated with major side effects and therapy resistance. Pelophen B (PPH), a synthetic analog of peloruside A, stabilizes microtubules through interaction with a non-taxoid binding site of β-tubulin. We evaluated the anticancer effect of PPH in a variety of tumor types by using established cell lines, early-passage cultures and ex vivo tumor-derived cultures that preserve the 3D architecture of the tumor microenvironment. PPH significantly blocks colony formation capacity, reduces viability and exerts additivity with PTX. Interestingly, PPH overcomes resistance to PTX. Mechanistically, PPH induces a G2/M cell cycle arrest and increases the presence of tubulin polymerization promoting protein (TPPP), inducing lysine 40 acetylation of α-tubulin. Although, results induced by paclitaxel or PPH are concordant, PPH's unique microtubule binding mechanism enables PTX additivity and ensures overcoming PTX-induced resistance. In conclusion, PPH results in remarkable anti-cancer activity in a range of preclinical models supporting further clinical investigation of PPH as a therapeutic anticancer agent.
Collapse
Affiliation(s)
- Stephanie Vermeulen
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | - Sam Ernst
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Experimental Surgery Lab, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Eva Blondeel
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | - Zihan Xia
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | - Pekka Rappu
- Department of Biochemistry, University of Turku, Turku, Finland
| | - Jyrki Heino
- Department of Biochemistry, University of Turku, Turku, Finland
| | - Sándor Dedeyne
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | - Hannelore Denys
- Cancer Research Institute Ghent, Ghent, Belgium
- Department of Medical Oncology, Ghent University Hospital, Ghent, Belgium
| | - Gwen Sys
- Cancer Research Institute Ghent, Ghent, Belgium
- Department of Orthopedic Surgery and Traumatology, Ghent University Hospital, Ghent, Belgium
| | - Stefanie Gijsels
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | - Herman Depypere
- Cancer Research Institute Ghent, Ghent, Belgium
- Department of Gynecology, Ghent University Hospital, Ghent, Belgium
| | - Philippe Tummers
- Cancer Research Institute Ghent, Ghent, Belgium
- Department of Gynecology, Ghent University Hospital, Ghent, Belgium
| | - Wim Ceelen
- Cancer Research Institute Ghent, Ghent, Belgium
- Experimental Surgery Lab, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | | | | | - Olivier Raes
- Department of Organic and Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - An Hendrix
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | - Johan Van der Eycken
- Department of Organic and Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Olivier De Wever
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Ghent, Belgium.
- Cancer Research Institute Ghent, Ghent, Belgium.
| |
Collapse
|
39
|
Bang S, Song JK, Lee KH. Hyperphosphorylated tau targeting human serum albumin Fusion protein as therapeutics for Alzheimer's diseases. IBRO Neurosci Rep 2024; 17:423-430. [PMID: 39634029 PMCID: PMC11615533 DOI: 10.1016/j.ibneur.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction Neurofibrillary tangles (NFTs) are composed of hyperphosphorylated forms of microtubule-associated protein tau (Tau), which is responsible for neurodegeneration in Alzheimer's disease (AD). The hippocampal region has been a major focus of AD research because the deposits of phosphorylated tau protein in these regions are correlated with early memory deficits. Despite extensive studies, therapeutic strategies to reduce tau hyperphosphorylation and NFTs deposition remain unclear. AL04, a recently developed recombinant fusion protein comprising Cystatin C, human serum albumin, and a novel blood brain barrier (BBB) penetrating peptide, is currently under investigation. Previous studies have demonstrated its effectiveness in reducing amyloid beta plaques in AD mouse model. Methods In this study, we investigated the effects of AL04 on lowering hyperphosphorylated tau and NFTs in JNPL3 mouse model harboring human tau-P301L mutation. 3-month-old female mice intraperitoneally received AL04 (5 mg/kg) or PBS treatment every other week for 24 weeks. We used confocal microscopy and western blot to visualize and analyze changes in hyperphosphorylated tau Ser202/Thr205 labeled with AT8 antibody in the brain. Results We found that the AL04 treatment decreases hyperphosphorylated tau at PP2A-sensitive epitope Ser202/Thr205 in the hippocampus of the brain. In the brain lysates of AL04 treated mice, we observed the reduction of I2PP2A, inhibitor of PP2A, and the induction of autophagy receptor proteins such as SQSTM-1/p62 and OPTN. Conclusion Our data suggests that AL04 can be used as an AD prophylactic/therapeutic agent as it lowers the hyperphosphorylated tau by downregulating I2PP2A. We also propose that AL04 can induce the degradation of hyperphosphorylated tau aggregates through the upregulation of the autophagy pathway.
Collapse
|
40
|
Mitroshina EV, Kalinina EP, Kalyakulina AI, Teplyakova AV, Vedunova MV. The Effect of the Optogenetic Stimulation of Astrocytes on Neural Network Activity in an In Vitro Model of Alzheimer's Disease. Int J Mol Sci 2024; 25:12237. [PMID: 39596305 PMCID: PMC11594756 DOI: 10.3390/ijms252212237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Optogenetics is a combination of optical and genetic technologies used to activate or, conversely, inhibit specific cells in living tissues. The possibilities of using optogenetics approaches for the treatment of epilepsy, Parkinson's and Alzheimer's disease (AD) are being actively researched. In recent years, it has become clear that one of the most important players in the development of AD is astrocytes. Astrocytes affect amyloid clearance, participate in the development of neuroinflammation, and regulate the functioning of neural networks. We used an adeno-associated virus carrying the glial fibrillary acidic protein (GFAP) promoter driving the optogenetic channelrhodopsin-2 (ChR2) gene to transduce astrocytes in primary mouse hippocampal cultures. We recorded the bioelectrical activity of neural networks from day 14 to day 21 of cultivation using multielectrode arrays. A single optogenetic stimulation of astrocytes at 14 day of cultivation (DIV14) did not cause significant changes in neural network bioelectrical activity. Chronic optogenetic stimulation from DIV14 to DIV21 exerts a stimulatory effect on the bioelectrical activity of primary hippocampal cultures (the proportion of spikes included in network bursts significantly increased since DIV19). Moreover, chronic optogenetic stimulation over seven days partially preserved the activity and functional architecture of neuronal network in amyloidosis modeling. These results suggest that the selective optogenetic activation of astrocytes may represent a promising novel therapeutic strategy for combating AD.
Collapse
Affiliation(s)
- Elena V. Mitroshina
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Avenue, 603022 Nizhny Novgorod, Russia
| | - Elizaveta P. Kalinina
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Avenue, 603022 Nizhny Novgorod, Russia
| | - Alena I. Kalyakulina
- Institute of Information Technologies, Mathematics and Mechanics, Lobachevsky State University, 603022 Nizhny Novgorod, Russia
- Institute of Biogerontology, Lobachevsky State University, 603022 Nizhny Novgorod, Russia
| | - Alexandra V. Teplyakova
- Federal Center of Brain Research and Neurotechnologies, Federal Medical-Biological Agency, 119330 Moscow, Russia
| | - Maria V. Vedunova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Avenue, 603022 Nizhny Novgorod, Russia
| |
Collapse
|
41
|
Zhong G, Chang X, Xie W, Zhou X. Targeted protein degradation: advances in drug discovery and clinical practice. Signal Transduct Target Ther 2024; 9:308. [PMID: 39500878 PMCID: PMC11539257 DOI: 10.1038/s41392-024-02004-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] [Received: 05/08/2024] [Revised: 08/19/2024] [Accepted: 09/28/2024] [Indexed: 11/08/2024] Open
Abstract
Targeted protein degradation (TPD) represents a revolutionary therapeutic strategy in disease management, providing a stark contrast to traditional therapeutic approaches like small molecule inhibitors that primarily focus on inhibiting protein function. This advanced technology capitalizes on the cell's intrinsic proteolytic systems, including the proteasome and lysosomal pathways, to selectively eliminate disease-causing proteins. TPD not only enhances the efficacy of treatments but also expands the scope of protein degradation applications. Despite its considerable potential, TPD faces challenges related to the properties of the drugs and their rational design. This review thoroughly explores the mechanisms and clinical advancements of TPD, from its initial conceptualization to practical implementation, with a particular focus on proteolysis-targeting chimeras and molecular glues. In addition, the review delves into emerging technologies and methodologies aimed at addressing these challenges and enhancing therapeutic efficacy. We also discuss the significant clinical trials and highlight the promising therapeutic outcomes associated with TPD drugs, illustrating their potential to transform the treatment landscape. Furthermore, the review considers the benefits of combining TPD with other therapies to enhance overall treatment effectiveness and overcome drug resistance. The future directions of TPD applications are also explored, presenting an optimistic perspective on further innovations. By offering a comprehensive overview of the current innovations and the challenges faced, this review assesses the transformative potential of TPD in revolutionizing drug development and disease management, setting the stage for a new era in medical therapy.
Collapse
Affiliation(s)
- Guangcai Zhong
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Xiaoyu Chang
- School of Pharmaceutical Sciences, Pingyuan Laboratory, Zhengzhou University, Zhengzhou, 450001, China
| | - Weilin Xie
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
| |
Collapse
|
42
|
Karakaya E, Abdul Y, Edwards J, Jamil S, Albayram O, Ergul A. Complex regulation of tau phosphorylation by the endothelin system in brain microvascular endothelial cells (BMVECs): link to barrier function. Clin Sci (Lond) 2024; 138:1329-1341. [PMID: 39356969 DOI: 10.1042/cs20240616] [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: 03/30/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/04/2024]
Abstract
Endothelin-1 (ET-1), the most potent vasoconstrictor identified to date, contributes to cerebrovascular dysfunction. ET-1 levels in postmortem brain specimens from individuals diagnosed with Alzheimer's disease (AD) and related dementias (ADRD) were shown to be related to cerebral hypoxia and disease severity. ET-1-mediated vascular dysfunction and ensuing cognitive deficits have also been reported in experimental models of AD and ADRD. Moreover, studies also showed that ET-1 secreted from brain microvascular endothelial cells (BMVECs) can affect neurovascular unit integrity in an autocrine and paracrine manner. Vascular contributions to cognitive impairment and dementia (VCID) is a leading ADRD cause known to be free of neuronal tau pathology, a hallmark of AD. However, a recent study reported cytotoxic hyperphosphorylated tau (p-tau) accumulation, which fails to bind or stabilize microtubules in BMVECs in VCID. Thus, the study aimed to determine the impact of ET-1 on tau pathology, microtubule organization, and barrier function in BMVECs. Cells were stimulated with 1 μM ET-1 for 24 h in the presence/absence of ETA (BQ123; 20 μM) or ETB (BQ788; 20 μM) receptor antagonists. Cell lysates were assayed for an array of phosphorylation site-specific antibodies and microtubule organization/stabilization markers. ET-1 stimulation increased p-tau Thr231 but decreased p-tau Ser199, Ser262, Ser396, and Ser214 levels only in the presence of ETA or ETB antagonism. ET-1 also impaired barrier function in the presence of ETA antagonism. These novel findings suggest that (1) dysregulation of endothelial tau phosphorylation may contribute to cerebral microvascular dysfunction and (2) the ET system may be an early intervention target to prevent hyperphosphorylated tau-mediated disruption of BMVEC barrier function.
Collapse
Affiliation(s)
- Eda Karakaya
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, U.S.A
- Ralph H. Johnson Veterans Affairs Health Care System, Charleston, SC, U.S.A
| | - Yasir Abdul
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, U.S.A
- Ralph H. Johnson Veterans Affairs Health Care System, Charleston, SC, U.S.A
| | - Jazlyn Edwards
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, U.S.A
| | - Sarah Jamil
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, U.S.A
- Ralph H. Johnson Veterans Affairs Health Care System, Charleston, SC, U.S.A
| | - Onder Albayram
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, U.S.A
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, U.S.A
| | - Adviye Ergul
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, U.S.A
- Ralph H. Johnson Veterans Affairs Health Care System, Charleston, SC, U.S.A
| |
Collapse
|
43
|
Odeh M, Sajrawi C, Majcher A, Zubedat S, Shaulov L, Radzishevsky A, Mizrahi L, Chung WK, Avital A, Hornemann T, Liebl DJ, Radzishevsky I, Wolosker H. A new type of blood-brain barrier aminoacidopathy underlies metabolic microcephaly associated with SLC1A4 mutations. Brain 2024; 147:3874-3889. [PMID: 38662784 PMCID: PMC11531853 DOI: 10.1093/brain/awae134] [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: 12/12/2023] [Revised: 03/19/2024] [Accepted: 04/05/2024] [Indexed: 10/20/2024] Open
Abstract
Mutations in the SLC1A4 transporter lead to neurodevelopmental impairments, spastic tetraplegia, thin corpus callosum and microcephaly in children. SLC1A4 catalyses obligatory amino acid exchange between neutral amino acids, but the physiopathology of SLC1A4 disease mutations and progressive microcephaly remain unclear. Here, we examined the phenotype and metabolic profile of three Slc1a4 mouse models: a constitutive Slc1a4-knockout mouse; a knock-in mouse with the major human Slc1a4 mutation (Slc1a4-K256E); and a selective knockout of Slc1a4 in brain endothelial cells (Slc1a4tie2-cre). We show that Slc1a4 is a bona fideL-serine transporter at the blood-brain barrier (BBB) and that acute inhibition or deletion of Slc1a4 leads to a decrease in serine influx into the brain. This results in microcephaly associated with decreased L-serine content in the brain, accumulation of atypical and cytotoxic 1-deoxysphingolipids, neurodegeneration, synaptic and mitochondrial abnormalities and behavioural impairments. Prenatal and early postnatal oral administration of L-serine at levels that replenish the serine pool in the brain rescued the observed biochemical and behavioural changes. Administration of L-serine until the second postnatal week also normalized brain weight in Slc1a4-E256K mice. Our observations suggest that the transport of 'non-essential' amino acids from the blood through the BBB is at least as important as that of essential amino acids for brain metabolism and development. We propose that SLC1A4 mutations cause a BBB aminoacidopathy with deficits in serine import across the BBB, required for optimal brain growth, leading to a metabolic microcephaly, which may be amenable to treatment with L-serine.
Collapse
Affiliation(s)
- Maali Odeh
- Department of Biochemistry, Rappaport Faculty of Medicine and Research Institute, Technion-Israel Inst of Technology, Haifa 3109601, Israel
| | - Clara Sajrawi
- Department of Biochemistry, Rappaport Faculty of Medicine and Research Institute, Technion-Israel Inst of Technology, Haifa 3109601, Israel
| | - Adam Majcher
- Institute of Clinical Chemistry, University of Zurich and University Hospital Zurich, Zurich 8091, Switzerland
| | - Salman Zubedat
- Department of Occupational Therapy, Faculty of Social Welfare and Health Sciences, University of Haifa, Haifa 3498838, Israel
| | - Lihi Shaulov
- Electron Microscopy Unit, B. Rappaport Faculty of Medicine, Technion-Israel Inst of Technology, Haifa 3109601, Israel
| | | | | | - Wendy K Chung
- Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Avi Avital
- Department of Occupational Therapy, Faculty of Social Welfare and Health Sciences, University of Haifa, Haifa 3498838, Israel
| | - Thorsten Hornemann
- Institute of Clinical Chemistry, University of Zurich and University Hospital Zurich, Zurich 8091, Switzerland
| | - Daniel J Liebl
- The Miami Project to Cure Paralysis, Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Inna Radzishevsky
- Department of Biochemistry, Rappaport Faculty of Medicine and Research Institute, Technion-Israel Inst of Technology, Haifa 3109601, Israel
| | - Herman Wolosker
- Department of Biochemistry, Rappaport Faculty of Medicine and Research Institute, Technion-Israel Inst of Technology, Haifa 3109601, Israel
- Laura and Isaac Perlmutter Metabolomics Center, Rappaport Faculty of Medicine and Research Institute, Technion-Israel Inst of Technology, Haifa 3109601, Israel
| |
Collapse
|
44
|
Aggidis A, Devitt G, Zhang Y, Chatterjee S, Townsend D, Fullwood NJ, Ortega ER, Tarutani A, Hasegawa M, Cooper A, Williamson P, Mendoza‐Oliva A, Diamond MI, Mudher A, Allsop D. A novel peptide-based tau aggregation inhibitor as a potential therapeutic for Alzheimer's disease and other tauopathies. Alzheimers Dement 2024; 20:7788-7804. [PMID: 39360630 PMCID: PMC11567856 DOI: 10.1002/alz.14246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/01/2024] [Accepted: 08/13/2024] [Indexed: 10/04/2024]
Abstract
INTRODUCTION As aggregation underpins Tau toxicity, aggregation inhibitor peptides may have disease-modifying potential. They are therefore currently being designed and target either the 306VQIVYK311 aggregation-promoting hotspot found in all Tau isoforms or the 275VQIINK280 aggregation-promoting hotspot found in 4R isoforms. However, for any Tau aggregation inhibitor to potentially be clinically relevant for other tauopathies, it should target both hotspots to suppress aggregation of Tau isoforms, be stable, cross the blood-brain barrier, and rescue aggregation-dependent Tau phenotypes in vivo. METHODS We developed a retro-inverso, stable D-amino peptide, RI-AG03 [Ac-rrrrrrrrGpkyk(ac)iqvGr-NH2], based on the 306VQIVYK311 hotspots which exhibit these disease-relevant attributes. RESULTS Unlike other aggregation inhibitors, RI-AG03 effectively suppresses aggregation of multiple Tau species containing both hotspots in vitro and in vivo, is non-toxic, and suppresses aggregation-dependent neurodegenerative and behavioral phenotypes. DISCUSSION RI-AG03 therefore meets many clinically relevant requirements for an anti-aggregation Tau therapeutic and should be explored further for its disease-modifying potential for Tauopathies. HIGHLIGHTS Our manuscript describes the development of a novel peptide inhibitor of Tau aggregation, a retro-inverso, stable D-amino peptide called RI-AG03 that displays many clinically relevant attributes. We show its efficacy in preventing Tau aggregation in both in vitro and in vivo experimental models while being non-toxic to cells. RI-AG03 also rescues a biosensor cell line that stably expresses Tau repeat domains with the P301S mutation fused to Cer/Clo and rescues aggregation-dependent phenotypes in vivo, suppressing neurodegeneration and extending lifespan. Collectively our data describe several properties and attributes of RI-AG03 that make it a promising disease-modifying candidate to explore for reducing pathogenic Tau aggregation in Tauopathies such as Alzheimer's disease. Given the real interest in reducing Tau aggregation and the potential clinical benefit of using such agents in clinical practice, RI-AG03 should be investigated further for the treatment of Tauopathies after validation in mammalian models. Tau aggregation inhibitors are the obvious first choice as Tau-based therapies as much of Tau-mediated toxicity is aggregation dependent. Indeed, there are many research efforts focusing on this therapeutic strategy with aggregation inhibitors being designed against one of the two aggregation-promoting hotspots of the Tau protein. To our knowledge, RI-AG03 is the only peptide aggregation inhibitor that inhibits aggregation of Tau by targeting both aggregation-promoting hotspot motifs simultaneously. As such, we believe that our study will have a significant impact on drug discovery efforts in this arena.
Collapse
Affiliation(s)
- Anthony Aggidis
- Department of Biological SciencesUniversity of SouthamptonSouthamptonUK
- Division of Biomedical and Life SciencesUniversity of LancasterLancasterUK
| | - George Devitt
- Department of Biological SciencesUniversity of SouthamptonSouthamptonUK
| | - Yongrui Zhang
- Department of Biological SciencesUniversity of SouthamptonSouthamptonUK
| | - Shreyasi Chatterjee
- Department of Biological SciencesUniversity of SouthamptonSouthamptonUK
- Department of Science and TechnologyNottingham Trent UniversityNottinghamUK
| | - David Townsend
- Department of ChemistryUniversity of LancasterLancaster UniversityLancasterUK
| | - Nigel J. Fullwood
- Division of Biomedical and Life SciencesUniversity of LancasterLancasterUK
| | - Eva Ruiz Ortega
- Department of Biological SciencesUniversity of SouthamptonSouthamptonUK
| | - Airi Tarutani
- Department of Dementia and Higher Brain FunctionTokyo Metropolitan Institute of Medical ScienceTokyoJapan
| | - Masato Hasegawa
- Department of Dementia and Higher Brain FunctionTokyo Metropolitan Institute of Medical ScienceTokyoJapan
| | - Amber Cooper
- Department of Biological SciencesUniversity of SouthamptonSouthamptonUK
| | - Philip Williamson
- Department of Biological SciencesUniversity of SouthamptonSouthamptonUK
| | - Ayde Mendoza‐Oliva
- Center for Alzheimer's and Neurodegenerative DiseasesPeter O'Donnell Jr. Brain InstituteUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Marc I. Diamond
- Center for Alzheimer's and Neurodegenerative DiseasesPeter O'Donnell Jr. Brain InstituteUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Amritpal Mudher
- Department of Biological SciencesUniversity of SouthamptonSouthamptonUK
| | - David Allsop
- Division of Biomedical and Life SciencesUniversity of LancasterLancasterUK
| |
Collapse
|
45
|
Yarbro JM, Han X, Dasgupta A, Yang K, Liu D, Shrestha HK, Zaman M, Wang Z, Yu K, Lee DG, Vanderwall D, Niu M, Sun H, Xie B, Chen PC, Jiao Y, Zhang X, Wu Z, Fu Y, Li Y, Yuan ZF, Wang X, Poudel S, Vagnerova B, He Q, Tang A, Ronaldson PT, Chang R, Yu G, Liu Y, Peng J. Human-mouse proteomics reveals the shared pathways in Alzheimer's disease and delayed protein turnover in the amyloidome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620263. [PMID: 39484428 PMCID: PMC11527136 DOI: 10.1101/2024.10.25.620263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Murine models of Alzheimer's disease (AD) are crucial for elucidating disease mechanisms but have limitations in fully representing AD molecular complexities. We comprehensively profiled age-dependent brain proteome and phosphoproteome (n > 10,000 for both) across multiple mouse models of amyloidosis. We identified shared pathways by integrating with human metadata, and prioritized novel components by multi-omics analysis. Collectively, two commonly used models (5xFAD and APP-KI) replicate 30% of the human protein alterations; additional genetic incorporation of tau and splicing pathologies increases this similarity to 42%. We dissected the proteome-transcriptome inconsistency in AD and 5xFAD mouse brains, revealing that inconsistent proteins are enriched within amyloid plaque microenvironment (amyloidome). Determining the 5xFAD proteome turnover demonstrates that amyloid formation delays the degradation of amyloidome components, including Aβ-binding proteins and autophagy/lysosomal proteins. Our proteomic strategy defines shared AD pathways, identify potential new targets, and underscores that protein turnover contributes to proteome-transcriptome discrepancies during AD progression.
Collapse
Affiliation(s)
- Jay M Yarbro
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- These authors contributed equally
| | - Xian Han
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- These authors contributed equally
| | - Abhijit Dasgupta
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Current address: Department of Computer Science and Engineering, SRM University AP, Andhra Pradesh 522240, India
- These authors contributed equally
| | - Ka Yang
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- These authors contributed equally
| | - Danting Liu
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Him K Shrestha
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Masihuz Zaman
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Zhen Wang
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Kaiwen Yu
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Dong Geun Lee
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - David Vanderwall
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Mingming Niu
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Huan Sun
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Boer Xie
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Ping-Chung Chen
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yun Jiao
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Xue Zhang
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Zhiping Wu
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yingxue Fu
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yuxin Li
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Zuo-Fei Yuan
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Xusheng Wang
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Suresh Poudel
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Barbora Vagnerova
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Qianying He
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Andrew Tang
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Patrick T Ronaldson
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Rui Chang
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Gang Yu
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yansheng Liu
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Cancer Research Institute, Yale University School of Medicine, West Haven, CT, 06516, USA
| | - Junmin Peng
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
46
|
Curley M, Rai M, Chuang CL, Pagala V, Stephan A, Coleman Z, Robles-Murguia M, Wang YD, Peng J, Demontis F. Transgenic sensors reveal compartment-specific effects of aggregation-prone proteins on subcellular proteostasis during aging. CELL REPORTS METHODS 2024; 4:100875. [PMID: 39383859 PMCID: PMC11573793 DOI: 10.1016/j.crmeth.2024.100875] [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: 12/14/2023] [Revised: 02/01/2024] [Accepted: 09/12/2024] [Indexed: 10/11/2024]
Abstract
Loss of proteostasis is a hallmark of aging that underlies many age-related diseases. Different cell compartments experience distinctive challenges in maintaining protein quality control, but how aging regulates subcellular proteostasis remains underexplored. Here, by targeting the misfolding-prone FlucDM luciferase to the cytoplasm, mitochondria, and nucleus, we established transgenic sensors to examine subcellular proteostasis in Drosophila. Analysis of detergent-insoluble and -soluble levels of compartment-targeted FlucDM variants indicates that thermal stress, cold shock, and pro-longevity inter-organ signaling differentially affect subcellular proteostasis during aging. Moreover, aggregation-prone proteins that cause different neurodegenerative diseases induce a diverse range of outcomes on FlucDM insolubility, suggesting that subcellular proteostasis is impaired in a disease-specific manner. Further analyses with FlucDM and mass spectrometry indicate that pathogenic tauV337M produces an unexpectedly complex regulation of solubility for different FlucDM variants and protein subsets. Altogether, compartment-targeted FlucDM sensors pinpoint a diverse modulation of subcellular proteostasis by aging regulators.
Collapse
Affiliation(s)
- Michelle Curley
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Mamta Rai
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Chia-Lung Chuang
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Vishwajeeth Pagala
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Anna Stephan
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Zane Coleman
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Maricela Robles-Murguia
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Yong-Dong Wang
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Junmin Peng
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA; Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA; Department of Structural Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Fabio Demontis
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA.
| |
Collapse
|
47
|
Vicidomini C, Fontanella F, D’Alessandro T, Roviello GN. A Survey on Computational Methods in Drug Discovery for Neurodegenerative Diseases. Biomolecules 2024; 14:1330. [PMID: 39456263 PMCID: PMC11506269 DOI: 10.3390/biom14101330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Currently, the age structure of the world population is changing due to declining birth rates and increasing life expectancy. As a result, physicians worldwide have to treat an increasing number of age-related diseases, of which neurological disorders represent a significant part. In this context, there is an urgent need to discover new therapeutic approaches to counteract the effects of neurodegeneration on human health, and computational science can be of pivotal importance for more effective neurodrug discovery. The knowledge of the molecular structure of the receptors and other biomolecules involved in neurological pathogenesis facilitates the design of new molecules as potential drugs to be used in the fight against diseases of high social relevance such as dementia, Alzheimer's disease (AD) and Parkinson's disease (PD), to cite only a few. However, the absence of comprehensive guidelines regarding the strengths and weaknesses of alternative approaches creates a fragmented and disconnected field, resulting in missed opportunities to enhance performance and achieve successful applications. This review aims to summarize some of the most innovative strategies based on computational methods used for neurodrug development. In particular, recent applications and the state-of-the-art of molecular docking and artificial intelligence for ligand- and target-based approaches in novel drug design were reviewed, highlighting the crucial role of in silico methods in the context of neurodrug discovery for neurodegenerative diseases.
Collapse
Affiliation(s)
- Caterina Vicidomini
- Institute of Biostructures and Bioimaging-Italian National Council for Research (IBB-CNR), Via De Amicis 95, 80145 Naples, Italy
| | - Francesco Fontanella
- Department of Electrical and Information Engineering “Maurizio Scarano”, University of Cassino and Southern Lazio, 03043 Cassino, Italy
| | - Tiziana D’Alessandro
- Department of Electrical and Information Engineering “Maurizio Scarano”, University of Cassino and Southern Lazio, 03043 Cassino, Italy
| | - Giovanni N. Roviello
- Institute of Biostructures and Bioimaging-Italian National Council for Research (IBB-CNR), Via De Amicis 95, 80145 Naples, Italy
| |
Collapse
|
48
|
Alshehri RS, Abuzinadah AR, Alrawaili MS, Alotaibi MK, Alsufyani HA, Alshanketi RM, AlShareef AA. A Review of Biomarkers of Amyotrophic Lateral Sclerosis: A Pathophysiologic Approach. Int J Mol Sci 2024; 25:10900. [PMID: 39456682 PMCID: PMC11507293 DOI: 10.3390/ijms252010900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/28/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by progressive degeneration of upper and lower motor neurons. The heterogeneous nature of ALS at the clinical, genetic, and pathological levels makes it challenging to develop diagnostic and prognostic tools that fit all disease phenotypes. Limitations associated with the functional scales and the qualitative nature of mainstay electrophysiological testing prompt the investigation of more objective quantitative assessment. Biofluid biomarkers have the potential to fill that gap by providing evidence of a disease process potentially early in the disease, its progression, and its response to therapy. In contrast to other neurodegenerative diseases, no biomarker has yet been validated in clinical use for ALS. Several fluid biomarkers have been investigated in clinical studies in ALS. Biofluid biomarkers reflect the different pathophysiological processes, from protein aggregation to muscle denervation. This review takes a pathophysiologic approach to summarizing the findings of clinical studies utilizing quantitative biofluid biomarkers in ALS, discusses the utility and shortcomings of each biomarker, and highlights the superiority of neurofilaments as biomarkers of neurodegeneration over other candidate biomarkers.
Collapse
Affiliation(s)
- Rawiah S. Alshehri
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia; (R.S.A.); (H.A.A.)
| | - Ahmad R. Abuzinadah
- Department of Neurology, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia; (M.S.A.); (A.A.A.)
- Neuromuscular Medicine Unit, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | - Moafaq S. Alrawaili
- Department of Neurology, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia; (M.S.A.); (A.A.A.)
- Neuromuscular Medicine Unit, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | - Muteb K. Alotaibi
- Neurology Department, Prince Sultan Military Medical City, Riyadh 12233, Saudi Arabia;
| | - Hadeel A. Alsufyani
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia; (R.S.A.); (H.A.A.)
| | - Rajaa M. Alshanketi
- Internal Medicine Department, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 22252, Saudi Arabia;
| | - Aysha A. AlShareef
- Department of Neurology, Faculty of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia; (M.S.A.); (A.A.A.)
- Neuromuscular Medicine Unit, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| |
Collapse
|
49
|
Capocchi JK, Figueroa-Romero C, Dunham SJB, Faraci G, Rothman JA, Whiteson KL, Seo DO, Holtzman DM, Grabrucker S, Nolan YM, Kaddurah-Daouk R, Jett DA. Symposium: What Does the Microbiome Tell Us about Prevention and Treatment of AD/ADRD? J Neurosci 2024; 44:e1295242024. [PMID: 39384409 PMCID: PMC11466070 DOI: 10.1523/jneurosci.1295-24.2024] [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: 07/07/2024] [Revised: 07/17/2024] [Accepted: 08/19/2024] [Indexed: 10/11/2024] Open
Abstract
Alzheimer's disease (AD) and Alzheimer's disease-related dementias (ADRDs) are broad-impact multifactorial neurodegenerative diseases. Their complexity presents unique challenges for developing effective therapies. This review highlights research presented at the 2024 Society for Neuroscience meeting which emphasized the gut microbiome's role in AD pathogenesis by influencing brain function and neurodegeneration through the microbiota-gut-brain axis. This emerging evidence underscores the potential for targeting the gut microbiota to treat AD/ADRD.
Collapse
Affiliation(s)
| | | | | | - Gina Faraci
- University of California, Irvine, Irvine, California 92697
| | - Jason A Rothman
- University of California, Irvine, Irvine, California 92697
- University of California, Riverside, Riverside, California 92521
| | | | - Dong-Oh Seo
- Washington University School of Medicine in St. Louis, St. Louis, Missouri 63110
| | - David M Holtzman
- Washington University School of Medicine in St. Louis, St. Louis, Missouri 63110
| | - Stefanie Grabrucker
- Department of Anatomy and Neuroscience, University College Cork, Cork T12 XF62, Ireland
| | - Yvonne M Nolan
- Department of Anatomy and Neuroscience, University College Cork, Cork T12 XF62, Ireland
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Ireland
| | | | - David A Jett
- National Institute of Neurological Disorders and Stroke, Rockville, Maryland 20852
| |
Collapse
|
50
|
Tang R, Franz CE, Hauger RL, Dale AM, Dorros SM, Eyler LT, Fennema-Notestine C, Hagler DJ, Lyons MJ, Panizzon MS, Puckett OK, Williams ME, Elman JA, Kremen WS. Early Cortical Microstructural Changes in Aging Are Linked to Vulnerability to Alzheimer's Disease Pathology. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2024; 9:975-985. [PMID: 38878863 PMCID: PMC11756816 DOI: 10.1016/j.bpsc.2024.05.012] [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: 02/21/2024] [Revised: 05/09/2024] [Accepted: 05/29/2024] [Indexed: 08/15/2024]
Abstract
BACKGROUND Early identification of Alzheimer's disease (AD) risk is critical for improving treatment success. Cortical thickness is a macrostructural measure used to assess neurodegeneration in AD. However, cortical microstructural changes appear to precede macrostructural atrophy and may improve early risk identification. Currently, whether cortical microstructural changes in aging are linked to vulnerability to AD pathophysiology remains unclear in nonclinical populations, who are precisely the target for early risk identification. METHODS In 194 adults, we calculated magnetic resonance imaging-derived maps of changes in cortical mean diffusivity (microstructure) and cortical thickness (macrostructure) over 5 to 6 years (mean age: time 1 = 61.82 years; time 2 = 67.48 years). Episodic memory was assessed using 3 well-established tests. We obtained positron emission tomography-derived maps of AD pathology deposition (amyloid-β, tau) and neurotransmitter receptors (cholinergic, glutamatergic) implicated in AD pathophysiology. Spatial correlational analyses were used to compare pattern similarity among maps. RESULTS Spatial patterns of cortical macrostructural changes resembled patterns of cortical organization sensitive to age-related processes (r = -0.31, p < .05), whereas microstructural changes resembled the patterns of tau deposition in AD (r = 0.39, p = .038). Individuals with patterns of microstructural changes that more closely resembled stereotypical tau deposition exhibited greater memory decline (β = 0.22, p = .029). Microstructural changes and AD pathology deposition were enriched in areas with greater densities of cholinergic and glutamatergic receptors (ps < .05). CONCLUSIONS Patterns of cortical microstructural changes were more AD-like than patterns of macrostructural changes, which appeared to reflect more general aging processes. Microstructural changes may better inform early risk prediction efforts as a sensitive measure of vulnerability to pathological processes prior to overt atrophy and cognitive decline.
Collapse
Affiliation(s)
- Rongxiang Tang
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California.
| | - Carol E Franz
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California
| | - Richard L Hauger
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California; Center of Excellence for Stress and Mental Health, Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Anders M Dale
- Department of Radiology, University of California San Diego, La Jolla, California; Department of Neurosciences, University of California San Diego, La Jolla, California
| | - Stephen M Dorros
- Department of Radiology, University of California San Diego, La Jolla, California
| | - Lisa T Eyler
- Department of Psychiatry, University of California San Diego, La Jolla, California; Desert Pacific Mental Illness Research Education and Clinical Center, Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Christine Fennema-Notestine
- Department of Psychiatry, University of California San Diego, La Jolla, California; Department of Radiology, University of California San Diego, La Jolla, California
| | - Donald J Hagler
- Department of Radiology, University of California San Diego, La Jolla, California
| | - Michael J Lyons
- Department of Psychological and Brain Sciences, Boston University, Boston, Massachusetts
| | - Matthew S Panizzon
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California
| | - Olivia K Puckett
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California
| | - McKenna E Williams
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California; Joint Doctoral Program in Clinical Psychology, San Diego State University/University of California San Diego, San Diego, California
| | - Jeremy A Elman
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California
| | - William S Kremen
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center for Behavior Genetics of Aging, University of California San Diego, La Jolla, California
| |
Collapse
|