101
|
Abdelhamed HG, Hassan AA, Sakraan AA, Al-Deeb RT, Mousa DM, Aboul Ezz HS, Noor NA, Khadrawy YA, Radwan NM. Brain interleukins and Alzheimer's disease. Metab Brain Dis 2025; 40:116. [PMID: 39891777 PMCID: PMC11787210 DOI: 10.1007/s11011-025-01538-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/10/2025] [Indexed: 02/03/2025]
Abstract
The central nervous system (CNS) is immune-privileged by several immuno-modulators as interleukins (ILs). ILs are cytokines secreted by immune cells for cell-cell signaling communications and affect the functions of the CNS. ILs were reported to orchestrate different molecular and cellular mechanisms of both physiological and pathological events, through overproduction or over-expression of their receptors. They interact with numerous receptors mediating pro-inflammatory and/or anti-inflammatory actions. Interleukins have been implicated to participate in neurodegenerative diseases. They play a critical role in Alzheimer's disease (AD) pathology which is characterized by the over-production of pro-inflammatory ILs. These may aggravate neurodegeneration, in addition to their contribution to detrimental mechanisms as oxidative stress, and excitotoxicity. However, recent research on the relation between ILs and AD revealed major discrepancies. Most of the major ILs were shown to play both pro- and anti-inflammatory roles in different experimental settings and models. The interactions between different ILs through shared pathways also add to the difficulty of drawing solid conclusions. In addition, targeting the different ILs has not yielded consistent results. The repeated failures of therapeutic drugs in treating AD necessitate the search for novel agents targeting multiple mechanisms of the disease pathology. In this context, the understanding of interleukins and their roles throughout the disease progression and interaction with other systems in the brain may provide promising therapeutic targets for the prevention or treatment of AD.
Collapse
Affiliation(s)
- Heba G Abdelhamed
- Department of Zoology and Chemistry, Faculty of Science, Cairo University, Giza, Egypt
| | - Arwa A Hassan
- Faculty of Pharmacy & Pharmaceutical Industries, Sinai University, Sinai, Egypt
| | - Alaa A Sakraan
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
| | | | - Dalia M Mousa
- Department of Biotechnology, Faculty of Science, Cairo University, Giza, Egypt
| | - Heba S Aboul Ezz
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt.
| | - Neveen A Noor
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
| | - Yasser A Khadrawy
- Medical Physiology Department, Medical Research and Clinical Studies Institute, National Research Center, Giza, Egypt
| | - Nasr M Radwan
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
102
|
Zhao H, Zhou X, Song Y, Zhao W, Sun Z, Zhu J, Yu Y. Multi-omics analyses identify gut microbiota-fecal metabolites-brain-cognition pathways in the Alzheimer's disease continuum. Alzheimers Res Ther 2025; 17:36. [PMID: 39893498 PMCID: PMC11786436 DOI: 10.1186/s13195-025-01683-0] [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: 05/06/2024] [Accepted: 01/22/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND Gut microbiota dysbiosis is linked to Alzheimer's disease (AD), but our understanding of the molecular and neuropathological bases underlying such association remains fragmentary. METHODS Using 16S rDNA amplicon sequencing, untargeted metabolomics, and multi-modal magnetic resonance imaging, we examined group differences in gut microbiome, fecal metabolome, neuroimaging measures, and cognitive variables across 30 patients with AD, 75 individuals with mild cognitive impairment (MCI), and 61 healthy controls (HC). Furthermore, we assessed the associations between these multi-omics changes using correlation and mediation analyses. RESULTS There were significant group differences in gut microbial composition, which were driven by 8 microbial taxa (e.g., Staphylococcus and Bacillus) exhibiting a progressive increase in relative abundance from HC to MCI to AD, and 2 taxa (e.g., Anaerostipes) showing a gradual decrease. 26 fecal metabolites (e.g., Arachidonic, Adrenic, and Lithocholic acids) exhibited a progressive increase from HC to MCI to AD. We also observed progressive gray matter atrophy in broadly distributed gray matter regions and gradual micro-structural integrity damage in widespread white matter tracts along the AD continuum. Integration of these multi-omics changes revealed significant associations between microbiota, metabolites, neuroimaging, and cognition. More importantly, we identified two potential mediation pathways: (1) microbiota → metabolites → neuroimaging → cognition, and (2) microbiota → metabolites → cognition. CONCLUSION Aside from elucidating the underlying mechanism whereby gut microbiota dysbiosis is linked to AD, our findings may contribute to groundwork for future interventions targeting the microbiota-metabolites-brain-cognition pathways as a therapeutic strategy in the AD continuum.
Collapse
Affiliation(s)
- Han Zhao
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Research Center of Clinical Medical Imaging, Anhui Province, Hefei, 230032, China
- Anhui Provincial Institute of Translational Medicine, Hefei, 230032, China
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, 230032, China
| | - Xia Zhou
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yu Song
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Research Center of Clinical Medical Imaging, Anhui Province, Hefei, 230032, China
- Anhui Provincial Institute of Translational Medicine, Hefei, 230032, China
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, 230032, China
| | - Wenming Zhao
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Research Center of Clinical Medical Imaging, Anhui Province, Hefei, 230032, China
- Anhui Provincial Institute of Translational Medicine, Hefei, 230032, China
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, 230032, China
| | - Zhongwu Sun
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| | - Jiajia Zhu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
- Research Center of Clinical Medical Imaging, Anhui Province, Hefei, 230032, China.
- Anhui Provincial Institute of Translational Medicine, Hefei, 230032, China.
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, 230032, China.
| | - Yongqiang Yu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
- Research Center of Clinical Medical Imaging, Anhui Province, Hefei, 230032, China.
- Anhui Provincial Institute of Translational Medicine, Hefei, 230032, China.
- Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei, 230032, China.
| |
Collapse
|
103
|
AmeliMojarad M, AmeliMojarad M, Cui X. An overview on the impact of viral pathogens on Alzheimer's disease. Ageing Res Rev 2025; 104:102615. [PMID: 39631533 DOI: 10.1016/j.arr.2024.102615] [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: 10/28/2024] [Revised: 11/30/2024] [Accepted: 11/30/2024] [Indexed: 12/07/2024]
Abstract
Alzheimer's disease (AD) is the most common type of dementia which affects over than 60 million cases worldwide with higher incidence in low and middle-income countries by 2030. Based on the multifactorial nature of AD different risk factors are linked to the condition considering the brain's β-amyloid plaques (Aβ) and neurofibrillary tangles (NFTs) as its primary hallmarks. Lately, viral photogenes specially after recent SARS-CoV-2 pandemic has gained a lot of attention in promoting the neurodegenerative disorder such as AD based on their capacity to increase the permeability of the blood-brain barrier, dysregulation of immune responses, and the impact on Aβ processing and phosphorylation of tau proteins. Therefore, in this review, we summarized the important association of viral pathogens and their mechanism by which they contribute with AD formation and development. AN OVERVIEW OF THE ROLES OF VIRAL PATHOGENS IN AD: According to this figure, viruses can infect neurons directly by modulating the BBB, transferring from endothelial cells to glial cells and then to neurons, increasing the Aβ deposition, and affecting the tau protein phosphorylation or indirectly through the virus's entrance and pathogenicity that can be accelerated by genetic and epigenetic factors, as well as chronic neuroinflammation caused by activated microglia and astrocytes.
Collapse
Affiliation(s)
- Melika AmeliMojarad
- Department of Oncology, The First Affiliate Hospital of Dalian Medical University, Zhongshan Road, Dalian, Liaoning 116011, China.
| | - Mandana AmeliMojarad
- Department of Oncology, The First Affiliate Hospital of Dalian Medical University, Zhongshan Road, Dalian, Liaoning 116011, China.
| | - Xiaonan Cui
- Department of Oncology, The First Affiliate Hospital of Dalian Medical University, Zhongshan Road, Dalian, Liaoning 116011, China.
| |
Collapse
|
104
|
Adeyemi OF, Gowland P, Bowtell R, Mougin O, Hosseini AA. Hippocampal Subfield Volume in Relation to Cerebrospinal Fluid Amyloid-ß in Early Alzheimer's Disease: Diagnostic Utility of 7T MRI. Eur J Neurol 2025; 32:e70076. [PMID: 39921301 PMCID: PMC11806197 DOI: 10.1111/ene.70076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 01/14/2025] [Accepted: 01/20/2025] [Indexed: 02/10/2025]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a neurodegenerative condition characterised by amyloid plaque accumulation and neurofibrillary tangles. Early detection is essential for effective intervention, but current diagnostic methods that enable early diagnosis in clinical practice rely on invasive or costly biomarker scanning. This study aimed to explore the utility of 7T MRI in assessing hippocampal subfield volumes and their correlation with cerebrospinal fluid (CSF) biomarkers in prodromal AD. METHODS Fifty-six participants, including AD patients and healthy controls, underwent 7T MRI scanning. Automated segmentation delineated hippocampal subfield volumes, with subsequent normalisation to whole brain volume. RESULTS Significant differences in hippocampal and subfield volumes were observed in prodromal AD patients, even when they did not exhibit high MTA scores on 3T MRI or show any whole brain volume loss. Additionally, the volume of the entorhinal cortex (ERC) correlated significantly with CSF amyloid-β levels, suggesting ERC's potential as a proxy CSF amyloid-ß measurement. Conversely, no significant associations were found between CSF 181-Phosphorylated-tau or total tau levels and any hippocampal subfield volumes. DISCUSSION These findings show the potential use of 7T MRI, particularly in ERC assessment, as a biomarker for early AD identification. Further validation studies are warranted to confirm these results and elucidate the relationship of ERC volume with CSF biomarkers.
Collapse
Affiliation(s)
- Oluwatobi F. Adeyemi
- Sir Peter Mansfield Imaging CentreUniversity of NottinghaNottinghamUK
- Department of PhysicsUniversity of AbujaAbujaNigeria
| | - Penny Gowland
- Sir Peter Mansfield Imaging CentreUniversity of NottinghaNottinghamUK
| | - Richard Bowtell
- Sir Peter Mansfield Imaging CentreUniversity of NottinghaNottinghamUK
| | - Olivier Mougin
- Sir Peter Mansfield Imaging CentreUniversity of NottinghaNottinghamUK
| | - Akram A. Hosseini
- Sir Peter Mansfield Imaging CentreUniversity of NottinghaNottinghamUK
- Department of Academic NeurologyNottingham University Hospitals NHS Trust, Queen's Medical CentreNottinghamUK
| |
Collapse
|
105
|
Wu MCS, Wei JHT, Fan RYS, Sim EZ, Yong KT, Gong T, Kong KV. Self-Assembled BODIPY@Au Core-Shell Structures for Durable Neuroprotective Phototherapy. Chembiochem 2025; 26:e202400562. [PMID: 39174489 DOI: 10.1002/cbic.202400562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 08/24/2024]
Abstract
BODIPY analogs are promising photosensitizers for molecular phototherapy; however, they exhibit high dark cytotoxicity and limited singlet oxygen generation capacity. In this study, we developed self-assembled core-shell nanophotosensitizers by linking a bipyridine group to BODIPY (Bpy-BODIPY) and promoting J-aggregation on gold nanourchins. This design enhances photostability and reduces the energy gap between the lowest singlet excited state and the lower triplet state, facilitating efficient singlet oxygen production. We characterized these nanophotosensitizers using UV-visible spectroscopy, transmission electron microscopy (TEM), surface-enhanced Raman spectroscopy (SERS) and dynamic light scattering (DLS), which confirmed the formation of the desired core-shell structure and J-aggregates. Notably, Bpy-BODIPY@Au significantly suppresses tau protein aggregation and enhances neuroprotective action, even in the presence of a phosphatase inhibitor. This work broadens the application of BODIPY chemistry to nanoagents for neuroprotective therapy.
Collapse
Affiliation(s)
- Melody Cai-Syaun Wu
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan R.O.C
| | - Jack Hau-Ting Wei
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan R.O.C
| | - Ricky Yu-Syun Fan
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan R.O.C
| | - Eng Zhi Sim
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan R.O.C
| | - Ken-Tye Yong
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, NSW, 2006, Australia
| | - Tianxun Gong
- School of Integrated Circuit Science and Engineering (Exemplary School of Microelectronics), University of Electronic Science and Technology of China, Chengdu, 611731, P. R. China
| | - Kien Voon Kong
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan R.O.C
| |
Collapse
|
106
|
Utpal BK, Roy SC, Zehravi M, Sweilam SH, Raja AD, Haque MA, Nayak C, Balakrishnan S, Singh LP, Panigrahi S, Alshehri MA, Rab SO, Minhaj NS, Emran TB. Polyphenols as Wnt/β-catenin pathway modulators: A promising strategy in clinical neurodegeneration. Animal Model Exp Med 2025; 8:266-286. [PMID: 39808166 PMCID: PMC11871115 DOI: 10.1002/ame2.12525] [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: 09/11/2024] [Accepted: 11/18/2024] [Indexed: 01/16/2025] Open
Abstract
Polyphenols, a diverse group of naturally occurring compounds found in plants, have garnered significant attention for their potential therapeutic properties in treating neurodegenerative diseases (NDs). The Wnt/β-catenin (WβC) signaling pathway, a crucial player in neurogenesis, neuronal survival, and synaptic plasticity, is involved in several cellular mechanisms related to NDs. Dysregulation of this pathway is a hallmark in the development of various NDs. This study explores multiple polyphenolic compounds, such as flavonoids, stilbenes, lignans, and phenolic acids, and their potential to protect the nervous system. It provides a comprehensive analysis of their effects on the WβC pathway, elucidating their modes of action. The study highlights the dual function of polyphenols in regulating and protecting the nervous system, providing reassurance about the research benefits. This review provides a comprehensive analysis of the results obtained from both in vitro studies and in vivo research, shedding light on how these substances influence the various components of the pathway. The focus is mainly on the molecular mechanisms that allow polyphenols to reduce oxidative stress, inflammation, and apoptotic processes, ultimately improving the function and survival of neurons. This study aims to offer a thorough understanding of the potential of polyphenols in targeting the WβC signaling pathway, which could lead to the development of innovative therapeutic options for NDs.
Collapse
Affiliation(s)
- Biswajit Kumar Utpal
- Department of Pharmacy, Faculty of Health and Life SciencesDaffodil International UniversityDhakaBangladesh
| | - Sajib Chandra Roy
- Department of Pharmacy, Faculty of PharmacyUniversity of DhakaDhakaBangladesh
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy, College of Dentistry and PharmacyBuraydah Private CollegesBuraydahSaudi Arabia
| | - Sherouk Hussein Sweilam
- Department of Pharmacognosy, College of PharmacyPrince Sattam Bin Abdulaziz UniversityAl‐KharjSaudi Arabia
- Department of Pharmacognosy, Faculty of PharmacyEgyptian Russian UniversityCairoEgypt
| | - A. Dinesh Raja
- Department of PharmaceuticsKMCH College of PharmacyCoimbatoreIndia
| | - M. Akiful Haque
- Department of Pharmaceutical Analysis, School of Pharmacy, Anurag University, HyderabadIndia
| | - Chandan Nayak
- Department of Pharmaceutics, School of PharmacyArka Jain UniversityJharkhandIndia
| | - Senthilkumar Balakrishnan
- Department of PharmaceuticsJKKMMRF‐Annai JKK Sampoorani Ammal College of PharmacyKomarapalayamNamakkalIndia
| | - Laliteshwar Pratap Singh
- Department of Pharmaceutical Chemistry, Narayan Institute of PharmacyGopal Narayan Singh UniversitySasaramIndia
| | - Saswati Panigrahi
- Department of Pharmaceutical ChemistrySt. John Institute of Pharmacy and ResearchVevoorPalgharIndia
| | | | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical ScienceKing Khalid UniversityAbhaSaudi Arabia
| | - Najmus Sakib Minhaj
- Department of Pharmacy, Faculty of PharmacyUniversity of DhakaDhakaBangladesh
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Health and Life SciencesDaffodil International UniversityDhakaBangladesh
| |
Collapse
|
107
|
Zhong X, Wang Q, Yang M, Lin G, Yao K, Wu Z, Xu D, Zhou H, Chen B, Shi H, Zhang M, Shi X, Zeng Y, Lao J, Liang S, Li J, Liu Q, Liu H, Chen Y, Lin Y, Ouyang C, Lv J, Liang X, Cheng Y, Ran P, Gong B, Zhang B, Guo J, Zhang H, Liu S, Zhang J, Liu H, Ning Y. Plasma p-tau217 and p-tau217/Aβ1-42 are effective biomarkers for identifying CSF- and PET imaging-diagnosed Alzheimer's disease: Insights for research and clinical practice. Alzheimers Dement 2025; 21:e14536. [PMID: 39887504 PMCID: PMC11848202 DOI: 10.1002/alz.14536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 12/06/2024] [Accepted: 12/18/2024] [Indexed: 02/01/2025]
Abstract
INTRODUCTION With the advancement of disease-modifying therapies for Alzheimer's disease (AD), validating plasma biomarkers against cerebrospinal fluid (CSF) and positron emission tomography (PET) standards is crucial in both research and real-world settings. METHODS We measured plasma phosphorylated tau (p-tau)217, p-tau181, amyloid beta (Aβ)1-40, Aβ1-42, and neurofilament light chain in research and real-world cohorts. Participants were categorized by brain amyloid status using US Food and Drug Administration/European Medicines Agency-approved CSF or PET methods. RESULTS Plasma p-tau217 and p-tau217/Aβ1-42 demonstrated superior accuracy in detecting brain amyloid pathologies, with area under the curve from 0.94 to 0.97 in all cohorts. Specificity was lower in the real-world cohort but improved significantly by integrating demographic and clinical factors, aligning performance with research cohorts. Additionally, plasma biomarkers exhibited strong correlations with their CSF counterparts and PET standardized uptake value ratios, with significant associations in amyloid-positive participants. DISCUSSION Plasma p-tau217 and p-tau217/Aβ1-42 are effective diagnostic tools. However, patient demographics, apolipoprotein E ε4 status, and cognitive condition must be considered to improve specificity in the clinical practice. HIGHLIGHTS Plasma phosphorylated tau (p-tau)217 and p-tau217/amyloid beta (Aβ)1-42 demonstrated exceptional accuracy (area under the curve: 0.94-0.97) in detecting brain amyloid pathologies across both research (Southern China Aging Brain Initiative [SCABI]-1, SCABI-2) and real-world clinical practice (RCP) cohorts. Incorporating patient-specific factors (sex, age, apolipoprotein E ε4, cognitive status) improved diagnostic specificity in the clinical RCP cohort, aligning its performance with that of research cohorts. Plasma biomarkers, particularly p-tau217 and their ratios, showed robust correlations with cerebrospinal fluid biomarkers and positron emission tomography amyloid standardized uptake value ratios, underscoring their value as non-invasive diagnostic alternatives. Plasma p-tau217 and p-tau217/Aβ1-42 proved highly effective in diagnosing amyloid burden, offering a practical solution to bridge research advancements with real-world clinical practice.
Collapse
|
108
|
Gao F, Zhan Y, Wang Q, Zhang M, Dai L, Shen Y. Pathological angiogenesis was associated with cerebrovascular lesion and neurodegeneration in Alzheimer's disease. Alzheimers Dement 2025; 21:e14521. [PMID: 39777972 PMCID: PMC11848169 DOI: 10.1002/alz.14521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/24/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025]
Abstract
INTRODUCTION We investigated the specific factors driving abnormal angiogenesis in Alzheimer's disease (AD) and its role in cerebrovascular lesions and neurodegeneration. METHODS We assessed cerebrovascular pathologies, amyloid-beta (Aβ), and tau pathologies in post mortem human brains and detected 12 angiogenic factors in cerebrospinal fluid (CSF) from the China Aging and Neurodegenerative Disease Initiative (CANDI) cohort. RESULTS We observed severe blood-brain barrier damage and elevated levels of the vascular marker CD31 in human AD brains, which had a stronger correlation with tau pathology than Aβ pathology. Consistently, only CSF pTau181 showed positive associations with CSF angiogenesis factors (soluble vascular endothelial growth factor receptor 2 [sVEGFR2], VEGF-C, VEGF-D, placental growth factor [PLGF], Angiopoietin2, and Serpin E1), but not CSF Aβ42/40. Additionally, higher levels of CSF sVEGFR1, soluble Tyrosine-protein kinase receptor 2 [sTIE2], PLGF, and interleukin 8 [IL8], as well as lower levels of CSF urokinase-type plasminogen activator [uPA], were associated with worsen cerebrovascular pathologies and neurodegeneration. DISCUSSION Our findings indicate that tau pathology may play a critical role in pathological angiogenesis, contributing to cerebrovascular lesions and neurodegeneration in AD. HIGHLIGHTS BBB damage and elevated vascular marker CD31 in human AD brains had a stronger correlation with tau pathology than Aβ pathology. CSF pTau181 mediated the effect of CSF Aβ42/40 on CSF sVEGFR1 and sTIE2. Only CSF pTau181 showed positive associations with sVEGFR2, VEGF-C, VEGF-D, PLGF, Angiopoietin2, and Serpin E1. Higher sVEGFR1, sTIE2, PLGF, and IL8, and lower uPA in CSF, were associated with cerebrovascular pathologies and neurodegeneration.
Collapse
Affiliation(s)
- Feng Gao
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Institute on Aging and Brain DisordersThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Anhui Province Key Laboratory of Biomedical Aging ResearchUniversity of Science and Technology of ChinaHefeiChina
| | - Yaxi Zhan
- Institute on Aging and Brain DisordersThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Qiong Wang
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Institute on Aging and Brain DisordersThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Anhui Province Key Laboratory of Biomedical Aging ResearchUniversity of Science and Technology of ChinaHefeiChina
| | - Mengguo Zhang
- Institute on Aging and Brain DisordersThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Linbin Dai
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Institute on Aging and Brain DisordersThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Anhui Province Key Laboratory of Biomedical Aging ResearchUniversity of Science and Technology of ChinaHefeiChina
| | - Yong Shen
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Institute on Aging and Brain DisordersThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Anhui Province Key Laboratory of Biomedical Aging ResearchUniversity of Science and Technology of ChinaHefeiChina
| |
Collapse
|
109
|
Wang Y, Wu W, Zeng F, Meng X, Peng M, Wang J, Chen Z, Liu W. The role of kynurenine pathway metabolism mediated by exercise in the microbial-gut-brain axis in Alzheimer's disease. Exp Neurol 2025; 384:115070. [PMID: 39603488 DOI: 10.1016/j.expneurol.2024.115070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
In recent years, the role of the microbiome-gut-brain axis in the pathogenesis of Alzheimer's disease (AD) has garnered increasing attention. Specifically, tryptophan metabolism via the kynurenine pathway (KP) plays a crucial regulatory role in this axis. This study reviews how exercise regulates the microbiome-gut-brain axis by influencing kynurenine pathway metabolism, thereby exerting resistance against AD. This paper also discusses how exercise positively impacts AD via the microbiome-gut-brain axis by modulating the endocrine, autonomic nervous, and immune systems. Although the specific mechanisms are not fully understood, research indicates that exercise may optimize tryptophan metabolism by promoting the growth of beneficial microbiota and inhibiting harmful microbiota, producing substances that are beneficial to the nervous system and combating AD. The aim of this review is to provide new perspectives and potential intervention strategies for the prevention and treatment of AD by exploring the links between exercise, KP and the gut-brain axis.
Collapse
Affiliation(s)
- Yiyang Wang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Weijia Wu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Fanqi Zeng
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Xiangyuan Meng
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Mei Peng
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Juan Wang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Zeyu Chen
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Wenfeng Liu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China; Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, Hunan Normal University, Changsha 410081, China.
| |
Collapse
|
110
|
Luo X, Liang J, Lei X, Sun F, Gong M, Liu B, Zhou Z. C/EBPβ in Alzheimer's disease: An integrative regulator of pathological mechanisms. Brain Res Bull 2025; 221:111198. [PMID: 39788461 DOI: 10.1016/j.brainresbull.2025.111198] [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: 10/24/2024] [Revised: 12/22/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
Alzheimer's disease (AD) stands as one of the most prevalent neurodegenerative disorders, characterized by a progressive decline in cognitive function, neuroinflammation, amyloid-beta (Aβ) plaques, and neurofibrillary tangles (NFTs). With the global aging population, the incidence of AD continues to rise, yet current therapeutic strategies remain limited in their ability to significantly alleviate cognitive impairments. Therefore, a deeper understanding of the molecular mechanisms underlying AD is imperative for the development of more effective treatments. In recent years, the transcription factor C/EBPβ has emerged as a pivotal regulator in several pathological processes of AD, including neuroinflammation, lipid metabolism, Aβ processing, and tau phosphorylation. Through intricate post-translational modifications, C/EBPβ modulates these processes and may influence the progression of AD on multiple fronts. This review systematically explores the multifaceted roles of C/EBPβ in the pathogenesis of AD, delving into its crucial involvement in neuroinflammation, Aβ production, tau pathology, and lipid metabolism dysregulation. Furthermore, we critically assess therapeutic strategies targeting C/EBPβ, examining the challenges and opportunities in regulating this factor. By synthesizing the latest research findings, we offer a more comprehensive understanding of the role of C/EBPβ in AD and discuss its potential as a therapeutic intervention target.
Collapse
Affiliation(s)
- Xiaoting Luo
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Junyi Liang
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Xue Lei
- The First Hospital Affiliated to Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Fengqi Sun
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | | | - Bin Liu
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China.
| | - Zhongguang Zhou
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| |
Collapse
|
111
|
Kanemaru E, Ichinose F. Essential role of sulfide oxidation in brain health and neurological disorders. Pharmacol Ther 2025; 266:108787. [PMID: 39719173 PMCID: PMC11806942 DOI: 10.1016/j.pharmthera.2024.108787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 11/21/2024] [Accepted: 12/19/2024] [Indexed: 12/26/2024]
Abstract
Hydrogen sulfide (H2S) is an environmental hazard well known for its neurotoxicity. In mammalian cells, H2S is predominantly generated by transsulfuration pathway enzymes. In addition, H2S produced by gut microbiome significantly contributes to the total sulfide burden in the body. Although low levels of H2S is believed to exert various physiological functions such as neurotransmission and vasomotor control, elevated levels of H2S inhibit the activity of cytochrome c oxidase (i.e., mitochondrial complex IV), thereby impairing oxidative phosphorylation. To protect the electron transport chain from respiratory poisoning by H2S, the compound is actively oxidized to form persulfides and polysulfides by a mitochondrial resident sulfide oxidation pathway. The reaction, catalyzed by sulfide:quinone oxidoreductase (SQOR), is the initial and critical step in sulfide oxidation. The persulfide species are subsequently oxidized to sulfite, thiosulfate, and sulfate by persulfide dioxygenase (ETHE1 or SDO), thiosulfate sulfurtransferase (TST), and sulfite oxidase (SUOX). While SQOR is abundantly expressed in the colon, liver, lung, and skeletal muscle, its expression is notably low in the brains of most mammals. Consequently, the brain's limited capacity to oxidize H2S renders it particularly sensitive to the deleterious effects of H2S accumulation. Impaired sulfide oxidation can lead to fatal encephalopathy, and the overproduction of H2S has been implicated in the developmental delays observed in Down syndrome. Our recent findings indicate that the brain's limited capacity to oxidize sulfide exacerbates its sensitivity to oxygen deprivation. The beneficial effects of sulfide oxidation are likely to be mediated not only by the detoxification of H2S but also by the formation of persulfide, which exerts cytoprotective effects through multiple mechanisms. Therefore, pharmacological agents designed to scavenge H2S and/or enhance persulfide levels may offer therapeutic potential against neurological disorders characterized by impaired or insufficient sulfide oxidation or excessive H2S production.
Collapse
Affiliation(s)
- Eiki Kanemaru
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| | - Fumito Ichinose
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
112
|
Wu G, Luo Y, Guo Q, Yang M, Mahaman YAR, Liu Y, Wang JZ, Liu R, Gao X, Wang X. Conformation pattern changes in R1-pS262 tau peptide induced endogenous tau aggregation, synaptic damage, and cognitive impairments. J Alzheimers Dis 2025; 103:951-965. [PMID: 39686621 DOI: 10.1177/13872877241307341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
BACKGROUND To date, the effect of tau phosphorylation at different amino acid sites on the conformation and function of tau is still unclear in Alzheimer's disease (AD). Protein fingerprinting, also known as the protein folding shape code (PFSC) method, is a protein structure prediction technique based on protein sequence, which can reveal proteins' most likely spatial conformation. OBJECTIVE To investigate the effect of phosphorylation on tau protein conformation using PFSC technology and further analyze the differences in the effect of phosphorylation on tau aggregation at specific sites. METHODS We performed a conformational analysis of wild-type and simulated mutant hTau441 using the PFSC method and synthesized the phosphorylated and non-phosphorylated tau fragments by the chemical solid phase method. RESULTS We found that the number of Ser262 protein fingerprints increased from six in tau S262A to nine in tau S262E, together with increased conformational changes and enhanced flexibility. The in vitro Thioflavin S assay showed that phosphorylated tau fragments R1-pS262 possessed a stronger activity of inducing tau aggregation. In contrast to the non-phosphorylated tau fragment R1-nS262, R1-pS262 promoted endogenous tau aggregation and decreased synaptic proteins. In rats, R1-pS262 caused cognitive impairments and neuronal loss in addition to endogenous tau aggregation and synaptic damage. CONCLUSIONS Our study firstly reports that tau phosphorylation at Ser262 induces tau aggregation, and phosphorylated tau fragments R1-pS262 directly result in neuropathological changes. These provide new clues to the pathogenesis of tauopathy, such as AD, and a new molecular target for possible intervention.
Collapse
Affiliation(s)
- Gang Wu
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yong Luo
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Qian Guo
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingming Yang
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yacoubou Abdoul Razak Mahaman
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Kidney Diseases, Medical College, Hubei Polytechnic University, Huangshi, China
| | - Yi Liu
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Zhi Wang
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Hubei Key Laboratory of Cognitive and Affective Disorders, School of Medicine, Jianghan University, Wuhan, China
| | - Rong Liu
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Gao
- Department of Pharmacy, Renmin Hospital, Wuhan University, Wuhan, China
| | - Xiaochuan Wang
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Hubei Key Laboratory of Cognitive and Affective Disorders, School of Medicine, Jianghan University, Wuhan, China
| |
Collapse
|
113
|
Chen YH, Wang ZB, Liu XP, Mao ZQ. Elevated serum sodium is linked to increased amyloid-dependent tau pathology, neurodegeneration, and cognitive impairment in Alzheimer's disease. J Neurochem 2025; 169:e16257. [PMID: 39503608 DOI: 10.1111/jnc.16257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 10/12/2024] [Accepted: 10/18/2024] [Indexed: 02/11/2025]
Abstract
Vascular dysfunction is implicated in the pathophysiology of Alzheimer's disease (AD). While sodium is essential for maintaining vascular function, its role in AD pathology remains unclear. We included 353 participants from the Alzheimer's Disease Neuroimaging Initiative (ADNI), assessing serum sodium levels, cerebrospinal fluid (CSF) and positron emission tomography (PET) biomarkers, magnetic resonance imaging (MRI), and cognitive function. An independent sample (N = 471) with available CSF sodium-related proteins and AD biomarkers was also included. Associations between serum sodium levels and AD pathology, neurodegeneration, and cognition were evaluated using linear regression models. Spearman's correlation analyses assessed the relationships between CSF sodium-related proteins and AD biomarkers. Higher serum sodium levels were associated with increased AD pathology, reduced hippocampal volume, and greater cognitive decline (all p < 0.05). The relationship between serum sodium and amyloid PET was evident in several AD-susceptible brain regions, including the neocortex and limbic system. Individuals with high serum sodium exhibited higher tau pathology, lower hippocampal volume, and more severe cognitive decline per unit increase in amyloid PET compared to those with low serum sodium (all p < 0.05). Among the 14 CSF sodium-related proteins, which were inter-correlated, six were significantly correlated with CSF AD pathology and amyloid PET, while two were correlated with hippocampal volume and cognitive function, with sodium channel subunit beta-2 (SCN2B) and sodium channel subunit beta-3 (SCN3B) showing the strongest correlations. These findings underscore the crucial role of serum sodium in AD progression, highlighting a potential network of sodium dysregulation involved in AD pathology. Targeting sodium may offer a novel therapeutic approach to slowing AD progression, particularly by impeding the progression of amyloid-related downstream events.
Collapse
Affiliation(s)
- Yu-Han Chen
- The First Clinical Medical School, Hebei North University, Zhangjiakou, China
| | - Zhi-Bo Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, China
| | - Xi-Peng Liu
- Department of Neurosurgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Zhi-Qi Mao
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
114
|
El-Desouky S, Abdel-Halim M, Fathalla RK, Abadi AH, Piazza GA, Salama M, El-Khodery SA, Youssef MA, Elfarrash S. A novel phosphodiesterase 5 inhibitor, RF26, improves memory impairment and ameliorates tau aggregation and neuroinflammation in the P301S tauopathy mouse model of Alzheimer's disease. Exp Neurol 2025; 384:115058. [PMID: 39549949 DOI: 10.1016/j.expneurol.2024.115058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/10/2024] [Accepted: 11/10/2024] [Indexed: 11/18/2024]
Abstract
Phosphodiesterase-5 (PDE5) inhibitors are primarily used in the treatment of erectile dysfunction and pulmonary hypertension, but have also been reported to have a potential therapeutic effect for the treatment of Alzheimer's disease (AD). This is likely to be through stimulation of nitric oxide (NO)/cyclic guanosine monophosphate (cGMP) signaling by elevating cGMP, a secondary messenger involved in processes of neuroplasticity. In the present study, we evaluated the efficacy of a novel PDE5 inhibitor, RF26, using P301S tauopathy mice model. A body of experimental evidence suggests that the development of tau inclusions leads to the neurodegeneration observed in tauopathies, including AD, Frontotemporal dementia (FTD), Supranuclear palsy and others. RF26 successfully targeted NO/cGMP signaling pathway and showed a significant improvement of spatial memory task performance of P301S mice using Morris Water Maze and T-maze. Furthermore, RF26 -treated mice showed a significant reduction of phosphorylated tau load, gliosis and downregulated pro-inflammatory cytokines. The presented data support the efficacy of RF26 as a potent PDE5 inhibitor and calls for further investigation as a potential therapeutic drug for Alzheimer's and other tauopathy related neurological disorders.
Collapse
Affiliation(s)
- Sara El-Desouky
- Medical experimental research center (MERC), Faculty of Medicine, Mansoura University, 35116 Mansoura, Egypt
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Reem K Fathalla
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo 11835, Egypt
| | - Gary A Piazza
- Department of Drug discovery and development, Harrison Collage of Pharmacy, Auburn University, Auburn, AL 36832, USA
| | - Mohamed Salama
- Institute of Global health and Human ecology, American University in Cairo, Egypt; Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Mansoura University, 35116 Mansoura, Egypt
| | - Sabry Ahmed El-Khodery
- Department of internal medicine, Faculty of Veterinary Medicine, Mansoura University, 35116 Mansoura, Egypt
| | - Mohamed A Youssef
- Department of internal medicine, Faculty of Veterinary Medicine, Mansoura University, 35116 Mansoura, Egypt
| | - Sara Elfarrash
- Medical experimental research center (MERC), Faculty of Medicine, Mansoura University, 35116 Mansoura, Egypt; Department of Medical Physiology, Faculty of Medicine, Mansoura University, 35116 Mansoura, Egypt.
| |
Collapse
|
115
|
Zheng Q, Wang X. Alzheimer's disease: insights into pathology, molecular mechanisms, and therapy. Protein Cell 2025; 16:83-120. [PMID: 38733347 PMCID: PMC11786724 DOI: 10.1093/procel/pwae026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024] Open
Abstract
Alzheimer's disease (AD), the leading cause of dementia, is characterized by the accumulation of amyloid plaques and neurofibrillary tangles in the brain. This condition casts a significant shadow on global health due to its complex and multifactorial nature. In addition to genetic predispositions, the development of AD is influenced by a myriad of risk factors, including aging, systemic inflammation, chronic health conditions, lifestyle, and environmental exposures. Recent advancements in understanding the complex pathophysiology of AD are paving the way for enhanced diagnostic techniques, improved risk assessment, and potentially effective prevention strategies. These discoveries are crucial in the quest to unravel the complexities of AD, offering a beacon of hope for improved management and treatment options for the millions affected by this debilitating disease.
Collapse
Affiliation(s)
- Qiuyang Zheng
- Shenzhen Research Institute of Xiamen University, Shenzhen 518057, China
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Xin Wang
- Shenzhen Research Institute of Xiamen University, Shenzhen 518057, China
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361005, China
| |
Collapse
|
116
|
Knezovic A, Salkovic-Petrisic M. Cholinergic neurotransmission in the brain of streptozotocin-induced rat model of sporadic Alzheimer's disease: long-term follow up. J Neural Transm (Vienna) 2025:10.1007/s00702-025-02887-2. [PMID: 39891708 DOI: 10.1007/s00702-025-02887-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 01/22/2025] [Indexed: 02/03/2025]
Abstract
Rats treated intracerebroventricularly with streptozotocin (STZ-icv) develop pathologic features, which resemble those in Alzheimer's disease and have been proposed as a non-transgenic model for sporadic type of the disease (sAD). We aimed to characterize cholinergic transmission in the rat brain as a function of STZ-icv dose and time after the treatment. Acetylcholinesterase (AChE) activity and expression of muscarinic (M1, M4) and nicotinic (α7) receptors, cholin acetyltransferase (ChAT) and glial fibrillary acidic protein (GFAP) were measured in hippocampus (HPC) and parietotemporal cortex (CTX) of STZ-icv and age-matched control rats one week, and one, three, six and nine months after the icv administration of STZ (0.3, 1 and 3 mg/kg), respectively. Cholinergic and astroglial changes were found most pronounced with a highest STZ dose in time-dependent manner. The cortex and hippocampus exhibited specific alterations in cholinergic transmission following STZ-icv administration, with either similar or distinct patterns depending on the parameter observed: increased AChE activity in HPC and invariable in CTX; increased M4 and ChAT levels in both regions; substantial cortical M1 level increment and moderate hippocampal M1 decrement; and decreased α7 levels in both regions, with subsequent increase observed only in HPC. Alterations in cerebral cholinergic neurotransmission in STZ-icv rat model were mostly following a threephasic time pattern: acute response (Phase I), complete/partial compensation (Phase II), and reappearance/progression of changes (Phase III). Staging structure of cholinergic changes in STZ-icv rat model might be speculated to partly correlate with cholinergic pathology in clinical AD stages.
Collapse
Affiliation(s)
- Ana Knezovic
- Department of Pharmacology, University of Zagreb School of Medicine, Salata 11, Zagreb, 10 000, Croatia.
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Salata 12, Zagreb, 10 000, Croatia.
| | - Melita Salkovic-Petrisic
- Department of Pharmacology, University of Zagreb School of Medicine, Salata 11, Zagreb, 10 000, Croatia
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Salata 12, Zagreb, 10 000, Croatia
| |
Collapse
|
117
|
Chen Z, Liu Y, Zhang Y, Zhu J, Li Q, Wu X. Enhanced Multimodal Low-Rank Embedding-Based Feature Selection Model for Multimodal Alzheimer's Disease Diagnosis. IEEE TRANSACTIONS ON MEDICAL IMAGING 2025; 44:815-827. [PMID: 39302791 DOI: 10.1109/tmi.2024.3464861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Identification of Alzheimer's disease (AD) with multimodal neuroimaging data has been receiving increasing attention. However, the presence of numerous redundant features and corrupted neuroimages within multimodal datasets poses significant challenges for existing methods. In this paper, we propose a feature selection method named Enhanced Multimodal Low-rank Embedding (EMLE) for multimodal AD diagnosis. Unlike previous methods utilizing convex relaxations of the -norm, EMLE exploits an -norm regularized projection matrix to obtain an embedding representation and select informative features jointly for each modality. The -norm, employing an upper-bounded nonconvex Minimax Concave Penalty (MCP) function to characterize sparsity, offers a superior approximation for the -norm compared to other convex relaxations. Next, a similarity graph is learned based on the self-expressiveness property to increase the robustness to corrupted data. As the approximation coefficient vectors of samples from the same class should be highly correlated, an MCP function introduced norm, i.e., matrix -norm, is applied to constrain the rank of the graph. Furthermore, recognizing that diverse modalities should share an underlying structure related to AD, we establish a consensus graph for all modalities to unveil intrinsic structures across multiple modalities. Finally, we fuse the embedding representations of all modalities into the label space to incorporate supervisory information. The results of extensive experiments on the Alzheimer's Disease Neuroimaging Initiative datasets verify the discriminability of the features selected by EMLE.
Collapse
|
118
|
Ayyanar MP, Vijayan M. A review on gut microbiota and miRNA crosstalk: implications for Alzheimer's disease. GeroScience 2025; 47:339-385. [PMID: 39562408 PMCID: PMC11872870 DOI: 10.1007/s11357-024-01432-5] [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: 11/07/2024] [Indexed: 11/21/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive decline and progressive neuronal damage. Recent research has highlighted the significant roles of the gut microbiota and microRNAs (miRNAs) in the pathogenesis of AD. This review explores the intricate interaction between gut microbiota and miRNAs, emphasizing their combined impact on Alzheimer's progression. First, we discuss the bidirectional communication within the gut-brain axis and how gut dysbiosis contributes to neuroinflammation and neurodegeneration in AD. Changes in gut microbiota composition in Alzheimer's patients have been linked to inflammation, which exacerbates disease progression. Next, we delve into the biology of miRNAs, focusing on their roles in gene regulation, neurodevelopment, and neurodegeneration. Dysregulated miRNAs are implicated in AD pathogenesis, influencing key processes like inflammation, tau pathology, and amyloid deposition. We then examine how the gut microbiota modulates miRNA expression, particularly in the brain, potentially altering neuroinflammatory responses and synaptic plasticity. The interplay between gut microbiota and miRNAs also affects blood-brain barrier integrity, further contributing to Alzheimer's pathology. Lastly, we explore therapeutic strategies targeting this gut microbiota-miRNA axis, including probiotics, prebiotics, and dietary interventions, aiming to modulate miRNA expression and improve AD outcomes. While promising, challenges remain in fully elucidating these interactions and translating them into effective therapies. This review highlights the importance of understanding the gut microbiota-miRNA relationship in AD, offering potential pathways for novel therapeutic approaches aimed at mitigating the disease's progression.
Collapse
Affiliation(s)
- Maruthu Pandian Ayyanar
- Department of Biology, The Gandhigram Rural Institute (Deemed to be University), Gandhigram, 624302, Tamil Nadu, India
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA.
| |
Collapse
|
119
|
Hoang LN, Lee H, Lee SJ. Improving cognitive impairment through chronic consumption of natural compounds/extracts: a systematic review and meta-analysis of randomized controlled trials. Front Aging Neurosci 2025; 16:1531278. [PMID: 39949865 PMCID: PMC11821934 DOI: 10.3389/fnagi.2024.1531278] [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: 11/20/2024] [Accepted: 12/30/2024] [Indexed: 02/16/2025] Open
Abstract
Introduction This systematic review and meta-analysis aimed to compare the efficacy of extended supplementation (≥6 weeks) with natural compounds or extracts in improving cognitive function in patients with mild cognitive impairment (MCI) or Alzheimer's disease (AD). Methods A comprehensive literature search was conducted across Cochrane, PubMed, PsycARTICLES, Scopus, and Web of Science databases from inception to April 10, 2024. Eligible studies were randomized controlled trials evaluating cognitive outcomes in patients with MCI or AD using the Mini-Mental State Examination (MMSE) and the Alzheimer's Disease Assessment Scale-Cognitive Subscale (ADAS-Cog). Results From an initial pool of 6,687 articles, 45 were deemed relevant for qualitative analysis. Of these, 37 studies demonstrated improvements or positive trends in cognitive outcomes with natural compound or extract supplementation. A total of 35 studies met the criteria for meta-analysis. The meta-analysis, involving 4,974 participants, revealed significant improvements in ADAS-Cog scores (pooled standardized mean difference = -2.88, 95% confidence interval [CI]: -4.26 to -1.50; t24 = -4.31, p < 0.01) following supplementation. Additionally, a suggestive trend toward improvement in MMSE scores was observed in a subgroup analysis of 1,717 participants (pooled standardized mean difference = 0.76, 95% CI: 0.06 to 1.46, t18 = 2.27, p = 0.04). Conclusion These findings support the potential cognitive benefits of extended (≥6 weeks) supplementation with natural compounds or extracts in individuals with MCI or AD. Further research is warranted to confirm these results and elucidate the underlying mechanisms. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/.
Collapse
Affiliation(s)
| | | | - Sook Jeong Lee
- Department of Bioactive Material Sciences and Research Centre of Bioactive Materials, Jeonbuk National University, Jeonju, Jeonbuk-do, Republic of Korea
| |
Collapse
|
120
|
Luo M, Zhu S, Dang H, Wen Q, Niu R, Long J, Wang Z, Tong Y, Ning Y, Yuan M, Xu G. Genetically-encoded targeted protein degradation technology to remove endogenous condensation-prone proteins and improve crop performance. Nat Commun 2025; 16:1159. [PMID: 39880812 PMCID: PMC11779824 DOI: 10.1038/s41467-025-56570-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: 09/13/2024] [Accepted: 01/22/2025] [Indexed: 01/31/2025] Open
Abstract
Effective modulation of gene expression in plants is achievable through tools like CRISPR and RNA interference, yet methods for directly modifying endogenous proteins remain lacking. Here, we identify the E3 ubiquitin ligase E3TCD1 and develope a Targeted Condensation-prone-protein Degradation (TCD) strategy. The X-E3TCD1 fusion protein acts as a genetically engineered degrader, selectively targeting endogenous proteins prone to condensation. For example, a transgenic E3TCD1 fusion with Teosinte branched 1 (TB1) degrades the native TB1 protein, resulting in increased tiller numbers in rice. Additionally, conditional degradation of the negative defense regulator Early Flowering 3 via a pathogen-responsive ProTBF1-uORFsTBF1 cassette enhances rice blast resistance without affecting flowering time in the absence of pathogen. Unlike prevailing targeted protein degradation strategies, the TCD system does not rely on small molecules, antibodies, or genetic knock-in fusion tags, demonstrating its promise as a transgene-based approach for optimizing crop performance.
Collapse
Affiliation(s)
- Ming Luo
- State Key Laboratory of Hybrid Rice, Institute for Advanced Studies (IAS), Wuhan University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, Hubei, China
- RNA Institute, Wuhan University, Wuhan, Hubei, China
| | - Sitao Zhu
- State Key Laboratory of Hybrid Rice, Institute for Advanced Studies (IAS), Wuhan University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, Hubei, China
- RNA Institute, Wuhan University, Wuhan, Hubei, China
| | - Hua Dang
- State Key Laboratory of Hybrid Rice, Institute for Advanced Studies (IAS), Wuhan University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, Hubei, China
- RNA Institute, Wuhan University, Wuhan, Hubei, China
| | - Qing Wen
- State Key Laboratory of Hybrid Rice, Institute for Advanced Studies (IAS), Wuhan University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, Hubei, China
- RNA Institute, Wuhan University, Wuhan, Hubei, China
| | - Ruixia Niu
- State Key Laboratory of Hybrid Rice, Institute for Advanced Studies (IAS), Wuhan University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, Hubei, China
- RNA Institute, Wuhan University, Wuhan, Hubei, China
| | - Jiawei Long
- State Key Laboratory of Hybrid Rice, Institute for Advanced Studies (IAS), Wuhan University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, Hubei, China
- RNA Institute, Wuhan University, Wuhan, Hubei, China
| | - Zhao Wang
- State Key Laboratory of Hybrid Rice, Institute for Advanced Studies (IAS), Wuhan University, Wuhan, Hubei, China
- Hubei Hongshan Laboratory, Wuhan, Hubei, China
- RNA Institute, Wuhan University, Wuhan, Hubei, China
| | - Yongjia Tong
- State Key Laboratory of Hybrid Rice, Institute for Advanced Studies (IAS), Wuhan University, Wuhan, Hubei, China
| | - Yuese Ning
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Meng Yuan
- Hubei Hongshan Laboratory, Wuhan, Hubei, China
- National Key Laboratory of Crop Genetic Improvement, National Centre of Plant Gene Research (Wuhan), Huazhong Agricultural University, Wuhan, Hubei, China
| | - Guoyong Xu
- State Key Laboratory of Hybrid Rice, Institute for Advanced Studies (IAS), Wuhan University, Wuhan, Hubei, China.
- Hubei Hongshan Laboratory, Wuhan, Hubei, China.
- RNA Institute, Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
121
|
Cruz-Sese J, Mirón-Alcala M, Alfonso-Triguero M, Olalde J, Ruiz L, Galbis-Gramage N, Cortes L, Escobar L, Preman P, Snellinx A, Saito T, Saido TC, Saiz-Aúz L, Rábano-Gutiérrez A, Tcw J, Goate A, Strooper BD, Alberdi E, Arranz AM. Divergent Effects of APOE3 and APOE4 Human Astrocytes on Key Alzheimer's Disease Hallmarks in Chimeric Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.28.635271. [PMID: 39975251 PMCID: PMC11838330 DOI: 10.1101/2025.01.28.635271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Despite strong evidence supporting that both astrocytes and apolipoprotein E (APOE) play crucial roles in the pathogenesis and progression of Alzheimer's disease (AD), the impact of astrocytes carrying different APOE variants on key AD pathological hallmarks remains largely unknown. To explore such effects in a human relevant context, we generated a chimeric model of AD. We transplanted isogenic APOE3 or APOE4 human induced pluripotent stem cell (hiPSC)-derived astrocyte progenitors into neonatal brains of AD model mice. We show that at five to six months after transplantation, transplanted cells have differentiated into mature astrocytes (h-astrocytes) that often integrate in upper layers of one cortical hemisphere. APOE3 and APOE4 h-astrocytes differentially express and secrete the APOE protein, which binds to Aβ plaques with an isoform-dependent affinity. Remarkably, APOE3 h-astrocytes ameliorate Aβ pathology, Tau pathology and neuritic dystrophy. In contrast, APOE4 h-astrocytes aggravate these AD processes. Moreover, APOE3 and APOE4 h-astrocytes modulate microglia responses to Aβ pathology in opposite ways. APOE4 h-astrocytes enhance microglia clustering around Aβ plaques and exacerbate DAM state whereas APOE3 h-astrocytes reduce microglia clustering and induce a more homeostatic state on plaque-associated microglia. These findings highlight a critical contribution of h-astrocytes not only to Aβ pathology but also to other key AD hallmarks in chimeric mice. In addition, our findings reveal that h-astrocytes with different APOE variants and the different forms of APOE they secrete have a crucial role in AD progression.
Collapse
|
122
|
Liu L, Zhang X, Chai Y, Zhang J, Deng Q, Chen X. Skull bone marrow and skull meninges channels: redefining the landscape of central nervous system immune surveillance. Cell Death Dis 2025; 16:53. [PMID: 39875352 PMCID: PMC11775313 DOI: 10.1038/s41419-025-07336-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 12/18/2024] [Accepted: 01/07/2025] [Indexed: 01/30/2025]
Abstract
The understanding of neuroimmune function has evolved from concepts of immune privilege and protection to a new stage of immune interaction. The discovery of skull meninges channels (SMCs) has opened new avenues for understanding central nervous system (CNS) immunity. Here, we characterize skull bone marrow and SMCs by detailing the anatomical structures adjacent to the skull, the differences between skull and peripheral bone marrow, mainstream animal processing methods, and the role of skull bone marrow in monitoring various CNS diseases. Additionally, we highlight several unresolved issues based on current research findings, aiming to guide future research directions.
Collapse
Affiliation(s)
- Liang Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, P.R. China
- Tianjin Neurological Institute, Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, P.R. China
| | - Xian Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, P.R. China
- Tianjin Neurological Institute, Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, P.R. China
| | - Yan Chai
- Tianjin Neurological Institute, Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, P.R. China
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, P.R. China
- Tianjin Neurological Institute, Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, P.R. China
| | - Quanjun Deng
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, P.R. China
| | - Xin Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, P.R. China.
- Tianjin Neurological Institute, Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, P.R. China.
| |
Collapse
|
123
|
Wang M, Hua Y, Bai Y. A review of the application of exercise intervention on improving cognition in patients with Alzheimer's disease: mechanisms and clinical studies. Rev Neurosci 2025; 36:1-25. [PMID: 39029521 DOI: 10.1515/revneuro-2024-0046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 07/08/2024] [Indexed: 07/21/2024]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, leading to sustained cognitive decline. An increasing number of studies suggest that exercise is an effective strategy to promote the improvement of cognition in AD. Mechanisms of the benefits of exercise intervention on cognitive function may include modulation of vascular factors by affecting cardiovascular risk factors, regulating cardiorespiratory health, and enhancing cerebral blood flow. Exercise also promotes neurogenesis by stimulating neurotrophic factors, affecting neuroplasticity in the brain. Additionally, regular exercise improves the neuropathological characteristics of AD by improving mitochondrial function, and the brain redox status. More and more attention has been paid to the effect of Aβ and tau pathology as well as sleep disorders on cognitive function in persons diagnosed with AD. Besides, there are various forms of exercise intervention in cognitive improvement in patients with AD, including aerobic exercise, resistance exercise, and multi-component exercise. Consequently, the purpose of this review is to summarize the findings of the mechanisms of exercise intervention on cognitive function in patients with AD, and also discuss the application of different exercise interventions in cognitive impairment in AD to provide a theoretical basis and reference for the selection of exercise intervention in cognitive rehabilitation in AD.
Collapse
Affiliation(s)
- Man Wang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, No. 12 Middle Wulumuqi Road, Jing'an District, Shanghai 200040, China
- Department of Rehabilitation Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yan Hua
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, No. 12 Middle Wulumuqi Road, Jing'an District, Shanghai 200040, China
| | - Yulong Bai
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, No. 12 Middle Wulumuqi Road, Jing'an District, Shanghai 200040, China
| |
Collapse
|
124
|
Guo MH, Lee WP, Vardarajan B, Schellenberg GD, Phillips-Cremins JE. Polygenic burden of short tandem repeat expansions promotes risk for Alzheimer's disease. Nat Commun 2025; 16:1126. [PMID: 39875385 PMCID: PMC11775329 DOI: 10.1038/s41467-025-56400-0] [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: 11/16/2023] [Accepted: 01/17/2025] [Indexed: 01/30/2025] Open
Abstract
Studies of the genetics of Alzheimer's disease (AD) have largely focused on single nucleotide variants and short insertions/deletions. However, most of the disease heritability has yet to be uncovered, suggesting that there is substantial genetic risk conferred by other forms of genetic variation. There are over one million short tandem repeats (STRs) in the genome, and their link to AD risk has not been assessed. As pathogenic expansions of STR cause over 30 neurologic diseases, it is important to ascertain whether STRs may also be implicated in AD risk. Here, we genotype 312,731 polymorphic STR tracts genome-wide using PCR-free whole genome sequencing data from 2981 individuals (1489 AD case and 1492 control individuals). We implement an approach to identify STR expansions as STRs with tract lengths that are outliers from the population. We then test for differences in aggregate burden of expansions in case versus control individuals. AD patients harbor a 1.19-fold increase of STR expansions compared to healthy elderly controls (p = 8.27×10-3, two-sided Mann-Whitney test). Individuals carrying >30 STR expansions have a 3.69-fold higher odds of having AD and have more severe AD neuropathology. AD STR expansions are highly enriched within active promoters in post-mortem hippocampal brain tissues and particularly within SINE-VNTR-Alu (SVA) retrotransposons. Together, these results demonstrate that expanded STRs within active promoter regions of the genome associate with risk of AD.
Collapse
Affiliation(s)
- Michael H Guo
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA.
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA.
| | - Wan-Ping Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Badri Vardarajan
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, USA
| | - Gerard D Schellenberg
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Jennifer E Phillips-Cremins
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA.
- Department of Bioengineering, University of Pennsylvania, Philadelphia, USA.
- Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA.
| |
Collapse
|
125
|
Le TTM, Trinh HTT, Pham HT, Nguyen DT, Do GH, Phan HT, Le TD, Quach NT, Phi TQ, Chu HH. Lycopodiaceae herb from Vietnam as a promising medicinal source of natural hupezine and novel huperzine-producing endophytic fungi. Biotechnol Lett 2025; 47:19. [PMID: 39856461 DOI: 10.1007/s10529-025-03562-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 10/07/2024] [Accepted: 01/10/2025] [Indexed: 01/27/2025]
Abstract
OBJECTIVES (1) To evaluate the potential of producing huperzine (Hup) and anticholinesterase (AChE) activities of nine native Lycopodiaceae species collected in Vietnam; (2) Isolation, identification and characterization of a novel fungus producing both HupA and HupB isolated from Lycopodium casuarinoides Spring. RESULTS All methanolic extracts of nine plants showed AChE inhibition from 8.55 to 71.81%. Of note, Huperzia serrata (Thunb.) Trevis, L. casuarinoides, Lycopodium clavatum L., Phlegmariurus squarrosus (G. Forst.), and P. phlegmaria (L.) T. Sen & U. Sen were shown to biosynthesize both HupA and HupB by high-performance liquid chromatography (HPLC). Plants H. serrata, L. casuarinoides and L. clavatum showed the most potent AchE IC50 inhibition. HupA and HupB concentrations from six plants were greater than those of previously reported Lycopodiaceae species. Sixty-four endophytic fungi were isolated from tissue of natural L. casuarinoides and then screened for HupA- and HupB-production by HPLC. Out of 64 fungal strains, only TTD2-2.7 extract could produce both HupA and HupB with the yields of 0.034 and 0.028 µg gdcw-1, respectively. Moreover, TTD2-2.7 extract also had inhibitory effects on AChE with the IC50 of 129.76 ± 4.13 µg ml-1, which was lower than the extract of host plant L. casuarinoides (94.03 ± 4.13 µg ml-1). The fungus was identified as Aspergillus sp. TTD2-2.7 by morphological characteristics and Internal Transcribed Spacer sequence analysis. CONCLUSIONS These are the first reports of (1) two species L. clavatum and L. casuarinoides producing both HupA and HupB, and (2) L. casuarinoides as novel sources of Hup-producing endophytic fungi as well as (3) fungus Aspergillus as a novel HupA- and HupB-producing endophyte isolated from L. casuarinoides.
Collapse
Affiliation(s)
- Thanh Thi Minh Le
- Institute of Biotechnology, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet, Cau Giay, Hanoi, 100000, Vietnam.
| | - Ha Thi Thu Trinh
- Institute of Biotechnology, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet, Cau Giay, Hanoi, 100000, Vietnam
| | - Ha Thanh Pham
- Institute of Biotechnology, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet, Cau Giay, Hanoi, 100000, Vietnam
| | - Dat Tien Nguyen
- Center of Research and Technology Transfer, VAST, 18 Hoang Quoc Viet, Cau Giay, Hanoi, 100000, Vietnam
| | - Giang Hoang Do
- Center of Research and Technology Transfer, VAST, 18 Hoang Quoc Viet, Cau Giay, Hanoi, 100000, Vietnam
| | - Hien Thuy Phan
- National Institute of Medicinal Materials, 3B Quang Trung, Hoan Kiem, Hanoi, 100000, Vietnam
| | - Tam Duc Le
- National Institute of Medicinal Materials, 3B Quang Trung, Hoan Kiem, Hanoi, 100000, Vietnam
| | - Ngoc Tung Quach
- Institute of Biotechnology, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet, Cau Giay, Hanoi, 100000, Vietnam
| | - Tien Quyet Phi
- Institute of Biotechnology, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet, Cau Giay, Hanoi, 100000, Vietnam
| | - Ha Hoang Chu
- Institute of Biotechnology, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet, Cau Giay, Hanoi, 100000, Vietnam
| |
Collapse
|
126
|
Wood JI, Dulewicz M, Ge J, Stringer K, Szadziewska A, Desai S, Koutarapu S, Hajar HB, Fenson L, Blennow K, Zetterberg H, Cummings DM, Savas JN, Edwards FA, Hanrieder J. Isotope Encoded Spatial Biology Identifies Amyloid Plaque-Age-Dependent Structural Maturation, Synaptic Loss, and Increased Toxicity. RESEARCH SQUARE 2025:rs.3.rs-5829037. [PMID: 39975899 PMCID: PMC11838767 DOI: 10.21203/rs.3.rs-5829037/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Understanding how amyloid beta (Aβ) plaques form and progress to neurotoxicity in Alzheimer's disease remains a significant challenge. This study aims to elucidate the processes involved in Aβ plaque formation and maturation using a knock-in Aβ mouse model (App NL-F/NL-F ). By employing mass spectrometry imaging and stable isotope labeling, we timestamped Aβ plaques from their initial deposition, enabling the spatial tracking of plaque aging. Correlating single-plaque spatial transcriptomics with time since seeding, allowed us to track gene-expression changes specifically associated with plaque age, independent of chronological age of the mouse or disease severity. We found that plaque age, within sections from individual mice aged from 10 to 18 months, negatively correlates with synaptic gene expression. Further, correlation with hyperspectral confocal microscopy using structure-specific dyes revealed a positive link between plaque age and structural maturity, with older plaques identified as more compact and associated with significantly greater synapse loss and toxicity.
Collapse
Affiliation(s)
- Jack I. Wood
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, House V, S-431 80 Mölndal, Sweden
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, United Kingdom
| | - Maciej Dulewicz
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, House V, S-431 80 Mölndal, Sweden
| | - Junyue Ge
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, House V, S-431 80 Mölndal, Sweden
| | - Katie Stringer
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, House V, S-431 80 Mölndal, Sweden
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, United Kingdom
| | - Alicja Szadziewska
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, House V, S-431 80 Mölndal, Sweden
| | - Sneha Desai
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, House V, S-431 80 Mölndal, Sweden
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, United Kingdom
| | - Srinivas Koutarapu
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, House V, S-431 80 Mölndal, Sweden
| | - Haady B. Hajar
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, United Kingdom
| | - Lydia Fenson
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, House V, S-431 80 Mölndal, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, House V, S-431 80 Mölndal, Sweden
- Clinical Neurochemistry LaboratoryMemory Clinic, Sahlgrenska University Hospital, Mölndal Hospital, House V, S-431 80 Mölndal, Sweden
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, P.R. China
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, House V, S-431 80 Mölndal, Sweden
- Clinical Neurochemistry LaboratoryMemory Clinic, Sahlgrenska University Hospital, Mölndal Hospital, House V, S-431 80 Mölndal, Sweden
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, Queens Square, WC1N 3BG London, United Kingdom
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, PR China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Damian M. Cummings
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, United Kingdom
| | - Jeffrey N. Savas
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Frances A. Edwards
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, United Kingdom
| | - Jörg Hanrieder
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal Hospital, House V, S-431 80 Mölndal, Sweden
- Clinical Neurochemistry LaboratoryMemory Clinic, Sahlgrenska University Hospital, Mölndal Hospital, House V, S-431 80 Mölndal, Sweden
- Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, Queens Square, WC1N 3BG London, United Kingdom
- Department of Neuropsychiatry, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
127
|
Kimura N, Sasaki K, Masuda T, Ataka T, Matsumoto M, Kitamura M, Nakamura Y, Matsubara E. Machine learning models for dementia screening to classify brain amyloid positivity on positron emission tomography using blood markers and demographic characteristics: a retrospective observational study. Alzheimers Res Ther 2025; 17:25. [PMID: 39838434 PMCID: PMC11748352 DOI: 10.1186/s13195-024-01650-1] [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: 09/26/2024] [Accepted: 12/17/2024] [Indexed: 01/23/2025]
Abstract
BACKGROUND Intracerebral amyloid β (Aβ) accumulation is considered the initial observable event in the pathological process of Alzheimer's disease (AD). Efficient screening for amyloid pathology is critical for identifying patients for early treatment. This study developed machine learning models to classify positron emission tomography (PET) Aβ-positivity in participants with preclinical and prodromal AD using data accessible to primary care physicians. METHODS This retrospective observational study assessed the classification performance of combinations of demographic characteristics, routine blood test results, and cognitive test scores to classify PET Aβ-positivity using machine learning. Participants with mild cognitive impairment (MCI) or normal cognitive function who visited Oita University Hospital or had participated in the USUKI study and met the study eligibility criteria were included. The primary endpoint was assessment of the classification performance of the presence or absence of intracerebral Aβ accumulation using five machine learning models (i.e., five combinations of variables), each constructed with three classification algorithms, resulting in a total of 15 patterns. L2-regularized logistic regression, and kernel Support Vector Machine (SVM) and Elastic Net algorithms were used to construct the classification models using 34 pre-selected variables (12 demographic characteristics, 11 blood test results, 11 cognitive test results). RESULTS Data from 262 records (260 unique participants) were analyzed. The mean (standard deviation [SD]) participant age was 73.8 (7.8) years. Using L2-regularized logistic regression, the mean receiver operating characteristic (ROC) area under the curve (AUC) (SD) in Model 0 (basic demographic characteristics) was 0.67 (0.01). Classification performance was similar in Model 1 (basic demographic characteristics and Mini Mental State Examination [MMSE] subscores) and Model 2 (demographic characteristics and blood test results) with a cross-validated mean ROC AUC (SD) of 0.70 (0.01) for both. Model 3 (demographic characteristics, blood test results, MMSE subscores) and Model 4 (Model 3 and ApoE4 phenotype) showed improved performance with a mean ROC AUC (SD) of 0.73 (0.01) and 0.76 (0.01), respectively. In models using blood test results, thyroid-stimulating hormone and mean corpuscular volume tended to be the largest contributors to classification. Classification performances were similar using the SVM and Elastic Net algorithms. CONCLUSIONS The machine learning models used in this study were useful for classifying PET Aβ-positivity using data from routine physician visits. TRIAL REGISTRATION UMIN Clinical Trials Registry (UMIN000051776, registered on 31/08/2023).
Collapse
Affiliation(s)
- Noriyuki Kimura
- Department of Neurology, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-machi, Yufu, Oita, 879-5593, Japan.
| | - Kotaro Sasaki
- Human Biology Integration Foundation, Deep Human Biology Learning, Eisai Co., Ltd, 4-6-10 Koishikawa, Bunkyo-ku, Tokyo, 112-8088, Japan.
| | - Teruaki Masuda
- Department of Neurology, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-machi, Yufu, Oita, 879-5593, Japan
| | - Takuya Ataka
- Department of Neurology, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-machi, Yufu, Oita, 879-5593, Japan
| | - Mariko Matsumoto
- Neurology Department, Medical Headquarters, Eisai Co., Ltd, 3-7-1 Nishi Shinjuku, Shinjuku-ku, Tokyo, 163-1023, Japan
| | - Mika Kitamura
- Neurology Department, Medical Headquarters, Eisai Co., Ltd, 3-7-1 Nishi Shinjuku, Shinjuku-ku, Tokyo, 163-1023, Japan
| | - Yosuke Nakamura
- Neurology Department, Medical Headquarters, Eisai Co., Ltd, 3-7-1 Nishi Shinjuku, Shinjuku-ku, Tokyo, 163-1023, Japan
| | - Etsuro Matsubara
- Department of Neurology, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-machi, Yufu, Oita, 879-5593, Japan
| |
Collapse
|
128
|
Flynn CM, Omoluabi T, Janes AM, Rodgers EJ, Torraville SE, Negandhi BL, Nobel TE, Mayengbam S, Yuan Q. Targeting early tau pathology: probiotic diet enhances cognitive function and reduces inflammation in a preclinical Alzheimer's model. Alzheimers Res Ther 2025; 17:24. [PMID: 39827356 PMCID: PMC11742226 DOI: 10.1186/s13195-025-01674-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) remains incurable, yet its long prodromal phase offers a crucial window for early intervention. Pretangle tau, a precursor to neurofibrillary tangles, plays a key role in early AD pathogenesis. Intervening in pretangle tau pathology could significantly delay the progression of AD. The gut-brain axis, increasingly recognized as a contributor to AD, represents a promising therapeutic target due to its role in regulating neuroinflammation and neurodegeneration. While probiotics have shown cognitive benefits in amyloid-centered AD models, their effect on pretangle tau pathology remains elusive. METHODS This study evaluates the effects of probiotics in a rat model of preclinical AD, specifically targeting hyperphosphorylated pretangle tau in the locus coeruleus. TH-CRE rats (N = 47; 24 females and 23 males) received either AAV carrying pseudophosphorylated human tau (htauE14) or a control virus at 3 months of age. Probiotic or control diets were administered at 9-12 months, with blood and fecal samples collected for ELISA and 16S rRNA gene sequencing. Behavioral assessments were conducted at 13-14 months, followed by analysis of brain inflammation, blood-brain barrier integrity, and GSK-3β activation. RESULTS Rats expressing pseudophosphorylated tau displayed impairment in spatial Y-maze (F1,39 = 4.228, p = 0.046), spontaneous object location (F1,39 = 6.240, p = 0.017), and olfactory discrimination (F1,39 = 7.521, p = 0.009) tests. Phosphorylation of tau at S262 (t3 = -4.834) and S356 (t3 = -3.258) in the locus coeruleus was parallelled by GSK-3β activation in the hippocampus (F1,24 = 10.530, p = 0.003). Probiotic supplementation increased gut microbiome diversity (F1,31 = 8.065, p = 0.007) and improved bacterial composition (F1,31 = 3.4867, p = 0.001). The enhancement in gut microbiomes was associated with enhanced spatial learning (p < 0.05), reduced inflammation indexed by Iba-1 (F1,25 = 5.284, p = 0.030) and CD-68 (F1,26 = 8.441, p = 0.007) expression, and inhibited GSK-3β in female rats (p < 0.01 compared to control females). CONCLUSIONS This study underscores the potential of probiotics to modulate the gut-brain axis and mitigate pretangle tau-related pathology in preclinical AD. Probiotic supplementation could offer a novel early intervention strategy for AD, highlighting the pivotal role of gut health in neurodegeneration.
Collapse
Affiliation(s)
- Cassandra M Flynn
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
| | - Tamunotonye Omoluabi
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
| | - Alyssa M Janes
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
- Biochemistry Department, Faculty of Science, Memorial University of Newfoundland, St. John's, NL, A1B 3X9, Canada
| | - Emma J Rodgers
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
- Psychology Department, Faculty of Science, Memorial University of Newfoundland, St. John's, NL, A1B 3X9, Canada
| | - Sarah E Torraville
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
| | - Brenda L Negandhi
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
| | - Timothy E Nobel
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada
| | - Shyamchand Mayengbam
- Biochemistry Department, Faculty of Science, Memorial University of Newfoundland, St. John's, NL, A1B 3X9, Canada
| | - Qi Yuan
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada.
| |
Collapse
|
129
|
Wei H, Du T, Zhang W, Ma W, Yao Y, Li J. Investigating the Therapeutic Mechanisms of Total Saikosaponins in Alzheimer's Disease: A Metabolomic and Proteomic Approach. Pharmaceuticals (Basel) 2025; 18:100. [PMID: 39861162 PMCID: PMC11768985 DOI: 10.3390/ph18010100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia among the elderly, yet effective treatments remain elusive. Total saikosaponins (TSS), the primary bioactive components in Bupleurum chinense, have shown promising therapeutic effects against AD in previous studies. Methods: To delve deeper into the mechanisms underlying the therapeutic role of TSS in AD, we investigated its neuroprotective effects and associated molecular mechanisms in APP/PS1 mice. Further, we employed metabolomic and proteomic analyses, with a focus on the potential protein-level changes induced by TSS, particularly those related to metabolite accumulation in the brain. Results: Our results showed that lysophosphatidylcholine, adenosine, and sphingomyelin in plasma might serve as potential biomarkers. Compared to the control group, AD mice exhibited significantly increased expression of proteins related to neuroinflammatory pathways, whereas proteins involved in cAMP signaling, cGMP-PKG signaling, and synaptic plasticity pathways were significantly downregulated. Notably, these signaling pathways were partially reversed in APP/PS1 mice following TSS administration. Behavioral tests demonstrated that TSS effectively improved the learning and memory functions of mice. Conclusions: Our findings suggest that TSS ameliorate cognitive decline through regulating neuroinflammatory pathways, cAMP and cGMP signaling, and synaptic plasticity pathways, providing insights into its therapeutic potential in AD.
Collapse
Affiliation(s)
- Huiling Wei
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China; (H.W.); (T.D.); (W.Z.); (W.M.)
| | - Tianyi Du
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China; (H.W.); (T.D.); (W.Z.); (W.M.)
| | - Weiwei Zhang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China; (H.W.); (T.D.); (W.Z.); (W.M.)
| | - Wei Ma
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China; (H.W.); (T.D.); (W.Z.); (W.M.)
| | - Yao Yao
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China; (H.W.); (T.D.); (W.Z.); (W.M.)
| | - Juan Li
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
- Ningxia Engineering and Technology Research Center for Modernization of Characteristic Chinese Medicine, and Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan 750004, China
| |
Collapse
|
130
|
Summat R, Waiwut P, Daodee S, Nualkaew N, Phemphunananchai K, Arsito PN, Chulikhit Y, Montakantirat O, Khamphukdee C, Boonyarat C. Phytomedicine Potential of Oroxylum indicum Root and Its Constituents: Targeting Alzheimer's Disease. PLANTS (BASEL, SWITZERLAND) 2025; 14:223. [PMID: 39861577 PMCID: PMC11769049 DOI: 10.3390/plants14020223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition characterized by a gradual decline in cognitive function, for which few effective treatments exist. This study investigated the neuroprotective potential of Oroxylum indicum root extract and its key constituents (baicalein, chrysin, oroxylin A) against AD hallmarks. The extract and its constituents exhibited antioxidant activity in the DPPH assay. They inhibited β-amyloid aggregation as measured by the thioflavin T assay and acetylcholinesterase activity using the Ellman method. In cell culture models, O. indicum extract showed an ability to protect neurons from the toxic effects of H2O2. Western blot analysis revealed the extract and its major active component, baicalein, downregulated pro-apoptotic markers (cleaved caspase-3, and BAX) upon H2O2 exposure. Furthermore, they reduced the expression of amyloidogenic proteins (BACE1) and phosphorylated tau. These findings suggest that O. indicum root extract, particularly baicalein, possesses multifaceted neuroprotective properties, targeting various aspects of AD pathogenesis, including oxidative stress, cholinergic dysfunction, β-amyloid formation, aggregation, and apoptosis. O. indicum root thus warrants further investigation as a promising source of therapeutic agents for AD.
Collapse
Affiliation(s)
- Rattana Summat
- Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (R.S.); (S.D.); (N.N.); (K.P.); (Y.C.); (O.M.); (C.K.)
| | - Pornthip Waiwut
- Faculty of Pharmaceutical Sciences, Ubon Ratchathani University, Ubon Ratchathani 34190, Thailand;
| | - Supawadee Daodee
- Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (R.S.); (S.D.); (N.N.); (K.P.); (Y.C.); (O.M.); (C.K.)
| | - Natsajee Nualkaew
- Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (R.S.); (S.D.); (N.N.); (K.P.); (Y.C.); (O.M.); (C.K.)
| | - Khemjira Phemphunananchai
- Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (R.S.); (S.D.); (N.N.); (K.P.); (Y.C.); (O.M.); (C.K.)
| | - Puguh Novi Arsito
- School of Pharmacy, Faculty of Medicine and Health Sciences, Universitas Muhammadiyah Yogyakarta, Yogyakarta 55183, Indonesia;
| | - Yaowared Chulikhit
- Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (R.S.); (S.D.); (N.N.); (K.P.); (Y.C.); (O.M.); (C.K.)
| | - Orawan Montakantirat
- Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (R.S.); (S.D.); (N.N.); (K.P.); (Y.C.); (O.M.); (C.K.)
| | - Charinya Khamphukdee
- Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (R.S.); (S.D.); (N.N.); (K.P.); (Y.C.); (O.M.); (C.K.)
| | - Chantana Boonyarat
- Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand; (R.S.); (S.D.); (N.N.); (K.P.); (Y.C.); (O.M.); (C.K.)
| |
Collapse
|
131
|
Matera A, Compagnion AC, Pedicone C, Kotah JM, Ivanov A, Monsorno K, Labouèbe G, Leggio L, Pereira-Iglesias M, Beule D, Mansuy-Aubert V, Williams TL, Iraci N, Sierra A, Marro SG, Goate AM, Eggen BJL, Kerr WG, Paolicelli RC. Microglial lipid phosphatase SHIP1 limits complement-mediated synaptic pruning in the healthy developing hippocampus. Immunity 2025; 58:197-217.e13. [PMID: 39657671 DOI: 10.1016/j.immuni.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 10/13/2024] [Accepted: 11/04/2024] [Indexed: 12/12/2024]
Abstract
The gene inositol polyphosphate-5-phosphatase D (INPP5D), which encodes the lipid phosphatase SH2-containing inositol polyphosphate 5-phosphatase 1 (SHIP1), is associated with the risk of Alzheimer's disease (AD). How it influences microglial function and brain physiology is unclear. Here, we showed that SHIP1 was enriched in early stages of healthy brain development. By combining in vivo loss-of-function approaches and proteomics, we discovered that mice conditionally lacking microglial SHIP1 displayed increased complement and synapse loss in the early postnatal brain. SHIP1-deficient microglia showed altered transcriptional signatures and abnormal synaptic pruning that was dependent on the complement system. Mice exhibited cognitive defects in adulthood only when microglial SHIP1 was depleted early postnatally but not at later stages. Induced pluripotent stem cell (iPSC)-derived microglia lacking SHIP1 also showed increased engulfment of synaptic structures. These findings suggest that SHIP1 is essential for proper microglia-mediated synapse remodeling in the healthy developing brain. Disrupting this process has lasting behavioral effects and may be linked to vulnerability to neurodegeneration.
Collapse
Affiliation(s)
- Alessandro Matera
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | | | - Chiara Pedicone
- Department of Genetics and Genomics Sciences, Icahn School of Medicine at Mt. Sinai, New York, NY, USA; Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Janssen M Kotah
- Department of Biomedical Sciences, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Andranik Ivanov
- Core Unit Bioinformatics, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Katia Monsorno
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Gwenaël Labouèbe
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Loredana Leggio
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Marta Pereira-Iglesias
- Achucarro Basque Center for Neuroscience, Barrio Sarriena s/n, Leioa, Spain; Department of Neuroscience, University of the Basque Country EHU/UPV, Barrio Sarriena s/n, Leioa, Spain
| | - Dieter Beule
- Core Unit Bioinformatics, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Tim L Williams
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Nunzio Iraci
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Amanda Sierra
- Achucarro Basque Center for Neuroscience, Barrio Sarriena s/n, Leioa, Spain; Department of Biochemistry and Molecular Biology, University of the Basque Country EHU/UPV, Barrio Sarriena, Leioa, Spain; Ikerbasque Foundation, Bilbao, Spain
| | - Samuele G Marro
- Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Neurosciences, Black Family Stem Cell Institute, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - Alison M Goate
- Department of Genetics and Genomics Sciences, Icahn School of Medicine at Mt. Sinai, New York, NY, USA; Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bart J L Eggen
- Department of Biomedical Sciences, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - William G Kerr
- Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA; Department of Pediatrics, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Rosa C Paolicelli
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
132
|
Zhang L, Zhou Y, Yang Z, Jiang L, Yan X, Zhu W, Shen Y, Wang B, Li J, Song J. Lipid droplets in central nervous system and functional profiles of brain cells containing lipid droplets in various diseases. J Neuroinflammation 2025; 22:7. [PMID: 39806503 PMCID: PMC11730833 DOI: 10.1186/s12974-025-03334-5] [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: 10/06/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Lipid droplets (LDs), serving as the convergence point of energy metabolism and multiple signaling pathways, have garnered increasing attention in recent years. Different cell types within the central nervous system (CNS) can regulate energy metabolism to generate or degrade LDs in response to diverse pathological stimuli. This article provides a comprehensive review on the composition of LDs in CNS, their generation and degradation processes, their interaction mechanisms with mitochondria, the distribution among different cell types, and the roles played by these cells-particularly microglia and astrocytes-in various prevalent neurological disorders. Additionally, we also emphasize the paradoxical role of LDs in post-cerebral ischemia inflammation and explore potential underlying mechanisms, aiming to identify novel therapeutic targets for this disease.
Collapse
Affiliation(s)
- Longxiao Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yunfei Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Zhongbo Yang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Liangchao Jiang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Xinyang Yan
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Wenkai Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yi Shen
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Bolong Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Jiaxi Li
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| | - Jinning Song
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
133
|
Shen Y, Zhang X, Liu S, Xin L, Xuan W, Zhuang C, Chen Y, Chen B, Zheng X, Wu R, Lin Y. CEST imaging combined with 1H-MRS reveal the neuroprotective effects of riluzole by improving neurotransmitter imbalances in Alzheimer's disease mice. Alzheimers Res Ther 2025; 17:20. [PMID: 39806490 PMCID: PMC11726951 DOI: 10.1186/s13195-025-01672-3] [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: 10/29/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025]
Abstract
BACKGROUND The imbalance of glutamate (Glu) and gamma-aminobutyric acid (GABA) neurotransmitter system plays a crucial role in the pathogenesis of Alzheimer's disease (AD). Riluzole is a Glu modulator originally approved for amyotrophic lateral sclerosis that has shown potential neuroprotective effects in various neurodegenerative disorders. However, whether riluzole can improve Glu and GABA homeostasis in AD brain and its related mechanism of action remain unknown. This study utilized chemical exchange saturation transfer (CEST) imaging combined with proton magnetic resonance spectroscopy (1H-MRS) to monitor the dynamic changes of Glu and GABA in riluzole-treated AD mice, aiming to evaluate the efficacy and mechanism of riluzole in AD treatment. METHODS GluCEST, GABACEST and 1H-MRS were used to longitudinally monitor Glu and GABA levels in 3xTg AD mice treated with riluzole (12.5 mg/kg/day) or vehicle for 20 weeks. Magnetic resonance measurements were performed at baseline, 6, 12, and 20 weeks post-treatment. Cognitive performance was assessed using the Morris Water Maze (MWM) at baseline, 10, and 20 weeks. At the study endpoint, immunohistochemistry, Nissl staining, and Western blot were used to evaluate the brain pathology, neuronal survival, and protein expression. RESULTS GluCEST, GABACEST and 1H-MRS consistently revealed higher levels of Glu and GABA in the brain of riluzole-treated AD mice compared to untreated controls, which were associated with improvements in spatial learning and memory. The cognitive improvements significantly correlated with the increased GluCEST signals and Glu levels. Immunohistochemistry and Nissl staining demonstrated that riluzole treatment reduced amyloid-beta (Aβ) deposition, tau hyperphosphorylation, GFAP-positive astrocyte activation, and prevented neuronal loss. Moreover, riluzole upregulated the expression of excitatory amino acid transporter 2 (EAAT2), glutamic acid decarboxylase 65/67 (GAD65/67), and glutamine synthetase (GS), suggesting enhanced neurotransmitter metabolism. CONCLUSIONS CEST imaging combined with 1H-MRS demonstrated the effectiveness of riluzole in modulating Glu- and GABA-related changes and improving cognitive function in 3xTg AD mice, potentially through regulating key proteins involved in neurotransmitter metabolism. These findings suggest riluzole as a therapeutic agent for Alzheimer's disease and highlight the utility of multimodal MR imaging in monitoring treatment response and exploring disease mechanisms.
Collapse
Affiliation(s)
- Yuanyu Shen
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Xiaolei Zhang
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Siqi Liu
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Lijing Xin
- Center for Biomedical Imaging (CIBM), Lausanne, Switzerland
- Animal Imaging and Technology, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Physics (IPHYS), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Wentao Xuan
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Caiyu Zhuang
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yue Chen
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Beibei Chen
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Xinhui Zheng
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Renhua Wu
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yan Lin
- Radiology Department, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China.
| |
Collapse
|
134
|
Jiang-Xie LF, Drieu A, Kipnis J. Waste clearance shapes aging brain health. Neuron 2025; 113:71-81. [PMID: 39395409 PMCID: PMC11717645 DOI: 10.1016/j.neuron.2024.09.017] [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: 08/15/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 10/14/2024]
Abstract
Brain health is intimately connected to fluid flow dynamics that cleanse the brain of potentially harmful waste material. This system is regulated by vascular dynamics, the maintenance of perivascular spaces, neural activity during sleep, and lymphatic drainage in the meningeal layers. However, aging can impinge on each of these layers of regulation, leading to impaired brain cleansing and the emergence of various age-associated neurological disorders, including Alzheimer's and Parkinson's diseases. Understanding the intricacies of fluid flow regulation in the brain and how this becomes altered with age could reveal new targets and therapeutic strategies to tackle age-associated neurological decline.
Collapse
Affiliation(s)
- Li-Feng Jiang-Xie
- Center for Brain Immunology and Glia (BIG), Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Antoine Drieu
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, 75014 Paris, France
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia (BIG), Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
135
|
Cruz E, Nisbet RM, Padmanabhan P, van Waardenberg AJ, Graham ME, Nkajja G, Tapaswi S, Connor BJ, Robinson P, Götz J. Proteostasis as a fundamental principle of Tau immunotherapy. Brain 2025; 148:168-184. [PMID: 39074206 PMCID: PMC11706327 DOI: 10.1093/brain/awae254] [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: 12/06/2023] [Revised: 05/01/2024] [Accepted: 07/08/2024] [Indexed: 07/31/2024] Open
Abstract
The microtubule-associated protein Tau is a driver of neuronal dysfunction in Alzheimer's disease and other tauopathies. In this process, Tau initially undergoes subtle changes to its abundance, subcellular localization and a vast array of post-translational modifications including phosphorylation that progressively result in the protein's somatodendritic accumulation and dysregulation of multiple Tau-dependent cellular processes. Given the various loss- and gain-of-functions of Tau in disease and the brain-wide changes in the proteome that characterize tauopathies, we asked whether targeting Tau would restore the alterations in proteostasis observed in disease. Therefore, by phage display, we generated a novel pan-Tau antibody, RNJ1, that preferentially binds human Tau and neutralizes proteopathic seeding activity in multiple cell lines and benchmarked it against a clinically tested pan-Tau antibody, HJ8.5 (murine version of tilavonemab). We then evaluated both antibodies, alone and in combination, in the K3 tauopathy mouse model, showing reduced Tau pathology and improvements in neuronal function following 14 weekly treatments, without obtaining synergy for the combination. These effects were more pronounced in female mice. To investigate the molecular mechanisms contributing to improvements in neuronal function, we employed quantitative proteomics, phosphoproteomics and kinase prediction analysis to first establish alterations in K3 mice relative to wild-type controls at the proteome level. In female K3 mice, we found 342 differentially abundant proteins, which are predominantly involved in metabolic and microtubule-associated processes, strengthening previously reported findings of defects in several functional domains in multiple tauopathy models. We next asked whether antibody-mediated Tau target engagement indirectly affects levels of deregulated proteins in the K3 model. Importantly, both immunotherapies, in particular RNJ1, induced abundance shifts towards a restoration to wild-type levels (proteostasis). A total of 257 of 342 (∼75%) proteins altered in K3 were closer in abundance to wild-type levels after RNJ1 treatment, and 73% after HJ8.5 treatment. However, the magnitude of these changes was less pronounced than that observed with RNJ1. Furthermore, analysis of the phosphoproteome showed an even stronger restoration effect with RNJ1, with ∼82% of altered phosphopeptides in K3 showing a shift to wild-type levels, and 75% with HJ8.5. Gene set over-representation analysis further confirmed that proteins undergoing restoration are involved in biological pathways affected in K3 mice. Together, our study suggests that a Tau immunotherapy-induced restoration of proteostasis links target engagement and treatment efficacy.
Collapse
Affiliation(s)
- Esteban Cruz
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, St Lucia Campus (Brisbane), Brisbane, QLD 4072, Australia
| | - Rebecca M Nisbet
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, St Lucia Campus (Brisbane), Brisbane, QLD 4072, Australia
- The Florey, The University of Melbourne, Parkville, Melbourne, VIC 3052, Australia
| | - Pranesh Padmanabhan
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, St Lucia Campus (Brisbane), Brisbane, QLD 4072, Australia
| | | | - Mark E Graham
- Synapse Proteomics, Children’s Medical Research Institute, The University of Sydney, Westmead, NSW 2145, Australia
| | - Godfrey Nkajja
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, St Lucia Campus (Brisbane), Brisbane, QLD 4072, Australia
| | - Swara Tapaswi
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, St Lucia Campus (Brisbane), Brisbane, QLD 4072, Australia
| | - Bradley J Connor
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, St Lucia Campus (Brisbane), Brisbane, QLD 4072, Australia
| | - Phil Robinson
- Cell Signalling Unit, Children's Medical Research Institute, The University of Sydney, Westmead, NSW 2145, Australia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, St Lucia Campus (Brisbane), Brisbane, QLD 4072, Australia
| |
Collapse
|
136
|
Zha X, Liu X, Wei M, Huang H, Cao J, Liu S, Bian X, Zhang Y, Xiao F, Xie Y, Wang W, Zhang C. Microbiota-derived lysophosphatidylcholine alleviates Alzheimer's disease pathology via suppressing ferroptosis. Cell Metab 2025; 37:169-186.e9. [PMID: 39510074 DOI: 10.1016/j.cmet.2024.10.006] [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: 10/27/2023] [Revised: 04/16/2024] [Accepted: 10/04/2024] [Indexed: 11/15/2024]
Abstract
Alzheimer's disease (AD) is a pervasive neurodegenerative disorder, and new approaches for its prevention and therapy are critically needed. Here, we elucidate a gut-microbiome-brain axis that offers actionable perspectives for achieving this objective. Using the 5xFAD mouse model, we identify increased Clostridium abundance and decreased Bacteroides abundance as key features associated with β-amyloid (Aβ) burden. Treatment with Bacteroides ovatus, or its associated metabolite lysophosphatidylcholine (LPC), significantly reduces Aβ load and ameliorates cognitive impairment. Mechanistically, LPC acts through the orphan receptor GPR119, inhibiting ACSL4 expression, thereby suppressing ferroptosis and ameliorating AD pathologies. Analysis of fecal and serum samples from individuals with AD also reveals diminished levels of Bacteroides and LPC. This study thus identifies a B.ovatus-triggered pathway regulating AD pathologies and indicates that the use of single gut microbiota, metabolite, or small molecule compound may complement current prevention and treatment approaches for AD.
Collapse
Affiliation(s)
- Xu Zha
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair & Beijing Key Laboratory for Tumor Invasion and Metastasis, Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China; State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
| | - Xicheng Liu
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair & Beijing Key Laboratory for Tumor Invasion and Metastasis, Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China; State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China.
| | - Mengping Wei
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair & Beijing Key Laboratory for Tumor Invasion and Metastasis, Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China
| | - Huanwei Huang
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair & Beijing Key Laboratory for Tumor Invasion and Metastasis, Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China
| | - Jiaqi Cao
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair & Beijing Key Laboratory for Tumor Invasion and Metastasis, Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China
| | - Shuo Liu
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair & Beijing Key Laboratory for Tumor Invasion and Metastasis, Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China
| | - Xiaomei Bian
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair & Beijing Key Laboratory for Tumor Invasion and Metastasis, Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China
| | - Yuting Zhang
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair & Beijing Key Laboratory for Tumor Invasion and Metastasis, Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China
| | - Fenyan Xiao
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair & Beijing Key Laboratory for Tumor Invasion and Metastasis, Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China
| | - Yuping Xie
- National Center for Protein Sciences Beijing, State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Wei Wang
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair & Beijing Key Laboratory for Tumor Invasion and Metastasis, Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.
| | - Chen Zhang
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair & Beijing Key Laboratory for Tumor Invasion and Metastasis, Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China; State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China; Chinese Institute for Brain Research, Beijing, China.
| |
Collapse
|
137
|
Chaulagain B, Singh J. Penetratin and Mannose-Functionalized Cannabidiol Lipid Nanoparticles Encapsulating the BDNF Gene Reduce Amyloid-Induced Inflammation. Mol Pharm 2025; 22:154-167. [PMID: 39588752 PMCID: PMC11874068 DOI: 10.1021/acs.molpharmaceut.4c00811] [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] [Indexed: 11/27/2024]
Abstract
Inflammation is emerging as a critical player in the disease progression of Alzheimer's disease (AD) by its interaction with amyloid beta plaques in a feed-forward loop. There is also a decline in the nourishment and enriching neurotrophic factor, brain-derived neurotrophic factor (BDNF), in the brain. Therefore, supplementing the brain with BDNF by gene delivery and delivering the anti-inflammatory agent, cannabidiol (CBD) in this case, to mitigate inflammation-induced disease cascade offers an attractive treatment strategy. To achieve the brain localization of CBD and pBDNF, lipid nanoparticles (LNPs) functionalized with mannose and penetratin were utilized. CBD and pBDNF were successfully encapsulated in the LNPs (more than 80%) with a size less than 180 nm, polydispersity index less than 0.25, and zeta potential of 23 mV. CBD was released from the formulation over a period of a week. The dual-functionalized LNPs demonstrated higher cellular uptake of CBD and expressed a significantly higher amount of BDNF (p-value <0.05) after transfection than their nonmodified counterparts in four brain cell lines, i.e., brain endothelial cells (b.END3), immortalized microglia cells (IMGs), primary astrocytes, and primary neurons. Similarly, the permeation of CBD through the dual-modified LNPs across the in vitro coculture blood-brain barrier model was significantly higher (p-value <0.05) compared to free CBD or nonfunctionalized nanoparticles. The LNPs demonstrated anti-inflammatory activity against lipopolysaccharides and human amyloid beta1-42 oligomer induction as they reduced the protein and mRNA expression of pro-inflammatory cytokines TNF-α (p < 0.05) and IL-1β (p < 0.05) in IMG cells. In summary, the penetratin and mannose-functionalized LNPs encapsulating CBD and pBDNF could serve as a promising therapy in AD, requiring further validation in animal models.
Collapse
Affiliation(s)
- Bivek Chaulagain
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND 58105, USA
| | - Jagdish Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND 58105, USA
| |
Collapse
|
138
|
Liu T, Ren C, Guo W, Zhang X, Li Y, Wang Y, Zhang Q, Chen B, Dai J, Yan XX, Zhang J, Huo L, Cui M. Synthesis and preclinical evaluation of diarylamine derivative as Tau-PET radiotracer for Alzheimer's Disease. Eur J Med Chem 2025; 281:117046. [PMID: 39536496 DOI: 10.1016/j.ejmech.2024.117046] [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: 09/21/2024] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
The presence of aggregated Tau in the brain is a dominant pathological hallmark of Tauopathies, particularly in Alzheimer's disease (AD). Therefore, developing ligands that can specifically and sensitively bind to Tau aggregates is essential for diagnosing and monitoring therapeutic interventions. In this study, we further investigated the structural optimization of the diarylamine skeleton, which exhibited promising binding characteristics and biological properties. We supplementarily explored the effects of the number and position of nitrogen atoms, types of heteroatoms and aromatic moieties, and radioactive positions on affinity for Tau. Through a structure-activity relationship (SAR) analysis based on 125I-labeled diarylamine derivatives, [125I]A6 was identified as a lead compound due to its desirable binding properties and ability to penetrate the brain, making it suitable for conversion into a18F-labeled PET tracer. Satisfactorily, [18F]FA1 fulfilled critical requirements as a Tau radiotracer, demonstrating high specificity and selectivity for Tau, a clean off-target profile against Aβ plaques and monoamine oxidase B (MAO-B), and favorable in vivo brain kinetics, as confirmed by dynamic PET studies in rodents and non-human primates.
Collapse
Affiliation(s)
- Tianqing Liu
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, 100875, China
| | - Chao Ren
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Wantong Guo
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, 100875, China
| | - Xiaojun Zhang
- Department of Nuclear Medicine, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yuying Li
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, 100875, China.
| | - Yan Wang
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Hunan, 410013, China
| | - Qilei Zhang
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Hunan, 410013, China
| | - Baian Chen
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, 100069, China; Department of Laboratory Animal Sciences, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Jiapei Dai
- Wuhan Institute for Neuroscience and Neuroengineering, South-Central Minzu University, Wuhan, 430074, China.
| | - Xiao-Xin Yan
- Department of Anatomy and Neurobiology, Central South University Xiangya School of Medicine, Hunan, 410013, China
| | - Jinming Zhang
- Department of Nuclear Medicine, Chinese PLA General Hospital, Beijing, 100853, China
| | - Li Huo
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Beijing, 100730, China.
| | - Mengchao Cui
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, 100875, China; Center for Advanced Materials Research, Beijing Normal University at Zhuhai, Zhuhai, 519087, China.
| |
Collapse
|
139
|
Li W, Chen Q, Peng C, Yang D, Liu S, Lv Y, Jiang L, Xu S, Huang L. Roles of the Receptor for Advanced Glycation End Products and Its Ligands in the Pathogenesis of Alzheimer's Disease. Int J Mol Sci 2025; 26:403. [PMID: 39796257 PMCID: PMC11721675 DOI: 10.3390/ijms26010403] [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: 12/11/2024] [Revised: 01/02/2025] [Accepted: 01/04/2025] [Indexed: 01/13/2025] Open
Abstract
The Receptor for Advanced Glycation End Products (RAGE), part of the immunoglobulin superfamily, plays a significant role in various essential functions under both normal and pathological conditions, especially in the progression of Alzheimer's disease (AD). RAGE engages with several damage-associated molecular patterns (DAMPs), including advanced glycation end products (AGEs), beta-amyloid peptide (Aβ), high mobility group box 1 (HMGB1), and S100 calcium-binding proteins. This interaction impairs the brain's ability to clear Aβ, resulting in increased Aβ accumulation, neuronal injury, and mitochondrial dysfunction. This further promotes inflammatory responses and oxidative stress, ultimately leading to a range of age-related diseases. Given RAGE's significant role in AD, inhibitors that target RAGE and its ligands hold promise as new strategies for treating AD, offering new possibilities for alleviating and treating this serious neurodegenerative disease. This article reviews the various pathogenic mechanisms of AD and summarizes the literature on the interaction between RAGE and its ligands in various AD-related pathological processes, with a particular focus on the evidence and mechanisms by which RAGE interactions with AGEs, HMGB1, Aβ, and S100 proteins induce cognitive impairment in AD. Furthermore, the article discusses the principles of action of RAGE inhibitors and inhibitors targeting RAGE-ligand interactions, along with relevant clinical trials.
Collapse
Affiliation(s)
- Wen Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (W.L.); (Q.C.); (C.P.); (D.Y.); (S.L.); (Y.L.); (L.J.)
| | - Qiuping Chen
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (W.L.); (Q.C.); (C.P.); (D.Y.); (S.L.); (Y.L.); (L.J.)
| | - Chengjie Peng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (W.L.); (Q.C.); (C.P.); (D.Y.); (S.L.); (Y.L.); (L.J.)
| | - Dan Yang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (W.L.); (Q.C.); (C.P.); (D.Y.); (S.L.); (Y.L.); (L.J.)
| | - Si Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (W.L.); (Q.C.); (C.P.); (D.Y.); (S.L.); (Y.L.); (L.J.)
| | - Yanwen Lv
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (W.L.); (Q.C.); (C.P.); (D.Y.); (S.L.); (Y.L.); (L.J.)
| | - Langqi Jiang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (W.L.); (Q.C.); (C.P.); (D.Y.); (S.L.); (Y.L.); (L.J.)
| | - Shijun Xu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (W.L.); (Q.C.); (C.P.); (D.Y.); (S.L.); (Y.L.); (L.J.)
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Lihua Huang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (W.L.); (Q.C.); (C.P.); (D.Y.); (S.L.); (Y.L.); (L.J.)
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu 611137, China
| |
Collapse
|
140
|
Rao A, Chen N, Kim MJ, Blumenfeld J, Yip O, Liang Z, Shostak D, Hao Y, Nelson MR, Koutsodendris N, Grone B, Ding L, Yoon SY, Arriola P, Zilberter M, Huang Y. Microglia depletion reduces human neuronal APOE4-related pathologies in a chimeric Alzheimer's disease model. Cell Stem Cell 2025; 32:86-104.e7. [PMID: 39500314 DOI: 10.1016/j.stem.2024.10.005] [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: 11/16/2023] [Revised: 07/17/2024] [Accepted: 10/04/2024] [Indexed: 11/13/2024]
Abstract
Despite strong evidence supporting the important roles of both apolipoprotein E4 (APOE4) and microglia in Alzheimer's disease (AD) pathogenesis, the effects of microglia on neuronal APOE4-related AD pathogenesis remain elusive. To examine such effects, we utilized microglial depletion in a chimeric model with induced pluripotent stem cell (iPSC)-derived human neurons in mouse hippocampus. Specifically, we transplanted homozygous APOE4, isogenic APOE3, and APOE-knockout (APOE-KO) iPSC-derived human neurons into the hippocampus of human APOE3 or APOE4 knockin mice and then depleted microglia in half of the chimeric mice. We found that both neuronal APOE and microglial presence were important for the formation of Aβ and tau pathologies in an APOE isoform-dependent manner (APOE4 > APOE3). Single-cell RNA sequencing analysis identified two pro-inflammatory microglial subtypes with elevated MHC-II gene expression enriched in chimeric mice with human APOE4 neuron transplants. These findings highlight the concerted roles of neuronal APOE, especially APOE4, and microglia in AD pathogenesis.
Collapse
Affiliation(s)
- Antara Rao
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Nuo Chen
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA
| | - Min Joo Kim
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Jessica Blumenfeld
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Oscar Yip
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Zherui Liang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - David Shostak
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Yanxia Hao
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA
| | - Maxine R Nelson
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Nicole Koutsodendris
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Brian Grone
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA
| | - Leo Ding
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA
| | - Seo Yeon Yoon
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
| | - Patrick Arriola
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
| | - Misha Zilberter
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
| | - Yadong Huang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA; Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA, USA; Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Department of Pathology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
141
|
Ferreira IL, Marinho D, de Rosa V, Castanheira B, Fang Z, Caldeira GL, Mota SI, Rego AC. Linking activation of synaptic NMDA receptors-induced CREB signaling to brief exposure of cortical neurons to oligomeric amyloid-beta peptide. J Neurochem 2025; 169:e16222. [PMID: 39263896 DOI: 10.1111/jnc.16222] [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: 02/19/2024] [Revised: 08/13/2024] [Accepted: 08/15/2024] [Indexed: 09/13/2024]
Abstract
Amyloid-beta peptide oligomers (AβO) have been considered "primum movens" for a cascade of events that ultimately cause selective neuronal death in Alzheimer's disease (AD). However, initial events triggered by AβO have not been clearly defined. Synaptic (Syn) N-methyl-d-aspartate receptors (NMDAR) are known to activate cAMP response element-binding protein (CREB), a transcriptional factor involved in gene expression related to cell survival, memory formation and synaptic plasticity, whereas activation of extrasynaptic (ESyn) NMDARs was linked to excitotoxic events. In AD brain, CREB phosphorylation/activation was shown to be altered, along with dyshomeostasis of intracellular Ca2+ (Ca2+ i). Thus, in this work, we analyze acute/early and long-term AβO-mediated changes in CREB activation involving Syn or ESyn NMDARs in mature rat cortical neurons. Our findings show that acute AβO exposure produce early increase in phosphorylated CREB, reflecting CREB activity, in a process occurring through Syn NMDAR-mediated Ca2+ influx. Data also demonstrate that AβO long-term (24 h) exposure compromises synaptic function related to Ca2+-dependent CREB phosphorylation/activation and nuclear CREB levels and related target genes, namely Bdnf, Gadd45γ, and Btg2. Data suggest a dual effect of AβO following early or prolonged exposure in mature cortical neurons through the activation of the CREB signaling pathway, linked to the activation of Syn NMDARs.
Collapse
Affiliation(s)
- I Luísa Ferreira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Daniela Marinho
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Valéria de Rosa
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Bárbara Castanheira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Zongwei Fang
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Gladys L Caldeira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Sandra I Mota
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - A Cristina Rego
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
142
|
Caruso A, Tommonaro G, Vassallo A, Paris D, Monné M, Catalano A, Sinicropi MS, Saturnino C. Imino and Thioureidic Derivatives as New Tools for Alzheimer's Disease: Preliminary Studies. Chem Biol Drug Des 2025; 105:e70049. [PMID: 39821693 DOI: 10.1111/cbdd.70049] [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: 10/29/2024] [Revised: 11/27/2024] [Accepted: 01/04/2025] [Indexed: 01/19/2025]
Abstract
Alzheimer's disease is a neurodegenerative chronic disease with a severe social and economic impact in the societies, which still lacks an efficient therapy. Several pathophysiological events (β-amyloid [Aβ] deposits, τ-protein aggregation, loss of cholinergic activity, and oxidative stress) occurs in the progression of the disease. Therefore, the search for efficient multi-targeted agents for the treatment of Alzheimer's disease becomes indispensable. In this paper we evaluated the AChE inhibition by Ellman's method and antioxidant activity by DPPH assay of nine synthetic compounds: two hydroxy-benzene derivatives (1 and 2), three bis-thioureidic derivatives (3-5), two imidazole derivatives (6 and 7), and two phenylacetamide derivatives (8 and 9). The compound 2, (3s,5s,7s)-adamantan-1-yl 4-(((E)-2,5-dihydroxybenzylidene)amino)benzoate, exhibited the best antioxidant activity (30.00 ± 1.05 μM eq Trolox) and compound 4 showed the highest AChE inhibition value (IC50 [μM] 8.40 ± 0.32). In the search for a compound showing combined activities (antioxidant and AChE inhibition), the compound 4, octane-1,8-diyl-bis-S-amidinothiourea dihydrobromide, (19.02 ± 1.52 μM eq Trolox; IC50 [μM] 8.40 ± 0.32) was chosen to carry out a molecular docking study. The results showed that compound 4 has the ability to bind the active site of acetylcholinesterase with considerable affinity (estimated binding energies of -8.5 kcal/mol). All data indicate that compound 4 has the potential to be further investigated as a possible candidate in the Alzheimer's disease treatment.
Collapse
Affiliation(s)
- Anna Caruso
- Department of Health Sciences, University of Basilicata, Potenza, Italy
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Italy
| | - Giuseppina Tommonaro
- National Council of Researches of Italy-Institute of Biomolecular Chemistry (ICB), Pozzuoli, Italy
| | - Antonio Vassallo
- Department of Health Sciences, University of Basilicata, Potenza, Italy
| | - Debora Paris
- National Council of Researches of Italy-Institute of Biomolecular Chemistry (ICB), Pozzuoli, Italy
| | - Magnus Monné
- Department of Health Sciences, University of Basilicata, Potenza, Italy
| | - Alessia Catalano
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Maria Stefania Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Italy
| | - Carmela Saturnino
- Department of Health Sciences, University of Basilicata, Potenza, Italy
| |
Collapse
|
143
|
Xiong W, She W, Liu Y, Zhou H, Wang X, Li F, Li R, Wang J, Qin D, Jing S, Duan X, Jiang C, Xu C, He Y, Wang Z, Ye Q. Clinical-grade human dental pulp stem cells improve adult hippocampal neural regeneration and cognitive deficits in Alzheimer's disease. Theranostics 2025; 15:894-914. [PMID: 39776809 PMCID: PMC11700856 DOI: 10.7150/thno.102315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Disrupted hippocampal functions and progressive neuronal loss represent significant challenges in the treatment of Alzheimer's disease (AD). How to achieve the improvement of pathological progression and effective neural regeneration to ameliorate the intracerebral dysfunctional environment and cognitive impairment is the goal of the current AD therapy. Methods: We examined the therapeutic potential of clinical-grade human derived dental pulp stem cells (hDPSCs) in cognitive function and neuropathology in AD. Specifically, we investigated the effect of neural crest-specific derived hDPSCs on endogenous neural regeneration and long-term efficacy following a single transplantation in the triple-transgenic mouse model (3xTg-AD). Results: Our research demonstrated that a single administration of clinical-grade hDPSCs yielded dramatic short-term therapeutic benefits (5 weeks) and sustained partial efficacy (6 months) with respect to improving cognitive impairment and delaying typical pathological progression in 3xTg-AD mice. Intriguingly, exogenous hDPSCs were robustly self-differentiated into newborn functional neurons in the hippocampus of 3xTg-AD mice. The foremost evidence is provided that hDPSCs promote endogenic neural regeneration by enhancing the activation of the Wnt/β-catenin pathway, which may contribute to stabilizing the hippocampal neural network to reverse memory deficits. Conclusion: These findings highlight the multifunctional potential of hDPSCs in AD treatment, which enhances cognition through alleviating neuropathology and providing neural regenerative driving force. Understanding these multiplicity effects is critical to advancing the clinical translation of stem cell-based therapies for AD.
Collapse
Affiliation(s)
- Wei Xiong
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wenting She
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Ye Liu
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Heng Zhou
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Xinxin Wang
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Fang Li
- Center for Neurodegenerative Disease Research, and Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ruohan Li
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Junnan Wang
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Dongdong Qin
- Center for Neurodegenerative Disease Research, and Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shuili Jing
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Xingxiang Duan
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Cailei Jiang
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Chun Xu
- Sydney School of Dentistry, The University of Sydney, Sydney, NSW, Australia
| | - Yan He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Zhihao Wang
- Center for Neurodegenerative Disease Research, and Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qingsong Ye
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
- Sydney School of Dentistry, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
144
|
dos Santos Ono RM, dos Santos Moreira NC, Carvalho I, Passos GA, Sakamoto-Hojo ET. Novel donepezil-tacrine hybrid (TAHB3) induces neurodifferentiation, neuroprotective effects, and activates the PI3K/AKT pathway on PC12 cells. J Alzheimers Dis Rep 2025; 9:25424823241309268. [PMID: 40034521 PMCID: PMC11864261 DOI: 10.1177/25424823241309268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 12/05/2024] [Indexed: 03/05/2025] Open
Abstract
Background Alzheimer's disease (AD) is a neurodegenerative disease characterized by the impairment of cognitive functions and neuronal loss. AD has no cure; current treatments like acetylcholinesterase inhibitors (AChEI) alleviate symptoms but do not halt disease progression. Objective We aimed to evaluate the effects and mechanisms of two novel AChEI hybrid compounds (TAHB3 and TA8Amino), regarding cytotoxicity, neuroprotection and neurodifferentiation in PC12 cells. Methods The effects of TAHB3 and TA8Amino on neurodifferentiation were analyzed on PC12 cells which were treated with AChEI compounds for seven days, following morphological, and quantitative analyses to calculate the differentiation percentages, neurite length, and protein expression. Regarding cytotoxicity and neuroprotection assays, PC12 cells were differentiated into mature neurons, then treated with TAHB3 or TA8Amino, following a posttreatment with H2O2 (an inducer of oxidative damage); the analyses were performed using the XTT assay and flow cytometry. Results The hybrid compound TAHB3 induced differentiation of PC12 cells, but TA8Amino did not cause the same effect. Both compounds did not show cytotoxic effects to PC12 cells and did not change the cell cycle progression, nor induce cell death. Only TAHB3 showed neuroprotective potential against induced-oxidative damage, and TAHB3 increased the levels of p-AKT, suggesting its action through the activation of the PI3K/AKT pathway. Conclusions Our results showed that TAHB3 can induce neurodifferentiation, besides a neuroprotective activity, indicating the potential of AChEI hybrid compounds as novel candidates to be explored for the establishment of novel therapeutic strategies for patients with AD.
Collapse
Affiliation(s)
| | | | - Ivone Carvalho
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Geraldo Aleixo Passos
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo-USP, Brazil
- Laboratory of Genetics and Molecular Biology, Department of Basic and Oral Biology, Ribeirão Preto School of Dentistry, University of São Paulo-USP, Brazil
| | - Elza Tiemi Sakamoto-Hojo
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo-USP, Brazil
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo-USP, Brazil
| |
Collapse
|
145
|
Zhou X, Cao H, Jiang Y, Chen Y, Zhong H, Fu WY, Lo RMN, Wong BWY, Cheng EYL, Mok KY, Kwok TCY, Mok VCT, Ip FCF, Miyashita A, Hara N, Ikeuchi T, Hardy J, Chen Y, Fu AKY, Ip NY. Transethnic analysis identifies SORL1 variants and haplotypes protective against Alzheimer's disease. Alzheimers Dement 2025; 21:e14214. [PMID: 39655505 PMCID: PMC11772736 DOI: 10.1002/alz.14214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 07/25/2024] [Accepted: 07/29/2024] [Indexed: 01/03/2025]
Abstract
INTRODUCTION The SORL1 locus exhibits protective effects against Alzheimer's disease (AD) across ancestries, yet systematic studies in diverse populations are sparse. METHODS Logistic regression identified AD-associated SORL1 haplotypes in East Asian (N = 5249) and European (N = 8588) populations. Association analysis between SORL1 haplotypes and AD-associated traits or plasma biomarkers was conducted. The effects of non-synonymous mutations were assessed in cell-based systems. RESULTS Protective SORL1 variants/haplotypes were identified in the East Asian and European populations. Haplotype Hap_A showed a strong protective effect against AD in East Asians, linked to less severe AD phenotypes, higher SORL1 transcript levels, and plasma proteomic changes. A missense variant within Hap_A, rs2282647-C allele, was linked to a lower risk of AD and decreased expression of a truncated SORL1 protein isoform. DISCUSSION Our transethnic analysis revealed key SORL1 haplotypes that exert protective effects against AD, suggesting mechanisms of the protective role of SORL1 in AD. HIGHLIGHTS We examined the AD-protective mechanisms of SORL1 in the general population across diverse ancestral backgrounds by jointly analyzing data from three East Asian cohorts (ie, mainland China, Hong Kong, and Japan) and a European cohort. Comparative analysis unveiled key ethnic-specific SORL1 genetic variants and haplotypes. Among these, the SORL1 minor haplotype, Hap_A, emerged as the primary AD-protective factor in East Asians. Hap_A exerts significant AD-protective effects in both APOE ε4 carriers and non-carriers. SORL1 haplotype Hap_A is associated with cognitive function, brain volume, and the activity of specific neuronal and immune-related pathways closely connected to AD risk. Protective variants within Hap_A are linked to increased SORL1 expression in human tissues. We identified an isoform-specific missense variant in Hap_A that modifies the function and levels of a truncated SORL1 protein isoform that is poorly investigated.
Collapse
Affiliation(s)
- Xiaopu Zhou
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience CenterThe Hong Kong University of Science and TechnologyHong KongChina
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug DevelopmentHKUST Shenzhen Research InstituteShenzhenGuangdongChina
| | - Han Cao
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience CenterThe Hong Kong University of Science and TechnologyHong KongChina
| | - Yuanbing Jiang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience CenterThe Hong Kong University of Science and TechnologyHong KongChina
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
| | - Yuewen Chen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience CenterThe Hong Kong University of Science and TechnologyHong KongChina
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug DevelopmentHKUST Shenzhen Research InstituteShenzhenGuangdongChina
- SIAT–HKUST Joint Laboratory for Brain ScienceShenzhenGuangdongChina
- The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of SciencesShenzhen–Hong Kong Institute of Brain Science – Shenzhen Fundamental Research InstitutionsShenzhenGuangdongChina
| | - Huan Zhong
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience CenterThe Hong Kong University of Science and TechnologyHong KongChina
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
| | - Wing Yu Fu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience CenterThe Hong Kong University of Science and TechnologyHong KongChina
| | - Ronnie Ming Nok Lo
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience CenterThe Hong Kong University of Science and TechnologyHong KongChina
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
| | - Bonnie Wing Yan Wong
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience CenterThe Hong Kong University of Science and TechnologyHong KongChina
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
| | - Elaine Yee Ling Cheng
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience CenterThe Hong Kong University of Science and TechnologyHong KongChina
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
| | - Kin Ying Mok
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience CenterThe Hong Kong University of Science and TechnologyHong KongChina
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug DevelopmentHKUST Shenzhen Research InstituteShenzhenGuangdongChina
- Department of Molecular NeuroscienceUCL Institute of NeurologyLondonUK
| | - Timothy C. Y. Kwok
- Therese Pei Fong Chow Research Centre for Prevention of Dementia, Division of Geriatrics, Department of Medicine and TherapeuticsThe Chinese University of Hong KongHong KongChina
| | - Vincent C. T. Mok
- Gerald Choa Neuroscience Centre, Lui Che Woo Institute of Innovative Medicine, Therese Pei Fong Chow Research Centre for Prevention of Dementia, Division of Neurology, Department of Medicine and TherapeuticsThe Chinese University of Hong KongHong KongChina
| | - Fanny C. F. Ip
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience CenterThe Hong Kong University of Science and TechnologyHong KongChina
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug DevelopmentHKUST Shenzhen Research InstituteShenzhenGuangdongChina
| | | | - Akinori Miyashita
- Department of Molecular Genetics, Brain Research InstituteNiigata UniversityNiigataJapan
| | - Norikazu Hara
- Department of Molecular Genetics, Brain Research InstituteNiigata UniversityNiigataJapan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research InstituteNiigata UniversityNiigataJapan
| | - John Hardy
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
- Department of Molecular NeuroscienceUCL Institute of NeurologyLondonUK
- Institute for Advanced StudyThe Hong Kong University of Science and TechnologyHong KongChina
| | - Yu Chen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience CenterThe Hong Kong University of Science and TechnologyHong KongChina
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug DevelopmentHKUST Shenzhen Research InstituteShenzhenGuangdongChina
- SIAT–HKUST Joint Laboratory for Brain ScienceShenzhenGuangdongChina
- The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of SciencesShenzhen–Hong Kong Institute of Brain Science – Shenzhen Fundamental Research InstitutionsShenzhenGuangdongChina
| | - Amy K. Y. Fu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience CenterThe Hong Kong University of Science and TechnologyHong KongChina
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug DevelopmentHKUST Shenzhen Research InstituteShenzhenGuangdongChina
- SIAT–HKUST Joint Laboratory for Brain ScienceShenzhenGuangdongChina
| | - Nancy Y. Ip
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience CenterThe Hong Kong University of Science and TechnologyHong KongChina
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug DevelopmentHKUST Shenzhen Research InstituteShenzhenGuangdongChina
- SIAT–HKUST Joint Laboratory for Brain ScienceShenzhenGuangdongChina
| |
Collapse
|
146
|
Ma Y, Xu D, Gan Y, Chen Z, Chen Y, Han X. Adverse outcome pathway of Alzheimer's disease-like changes resulting from autophagy flux blockade after MC-LR exposure. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 364:125322. [PMID: 39549990 DOI: 10.1016/j.envpol.2024.125322] [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: 10/08/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 11/18/2024]
Abstract
Microcystins (MCs) pollution is a worldwide environmental issue concerning about human health. Microcystin-leucine-arginine (MC-LR), the most common type of MCs produced by cyanobacteria, could enter the brain and bring about damage to the nervous system. Up to date, it is not clear about the mechanism of MC-LR-induced neurotoxicity. Amyloid-β (Aβ) deposits are hallmark of Alzheimer's disease (AD). In this study, we revealed that MC-LR exposure at environment-related doses (1, 7.5, 15 μg/L) could promote Aβ accumulation in mouse brain. Mechanically, we firstly found that Aβ accumulation is closely associated with abnormal Aβ degradation due to autophagy flux blockade and lysosome dysfunctions in neurons after MC-LR exposure. Moreover, an adverse outcome pathway (AOP) framework oriented to neurotoxicity of MC-LR was conducted in this study. MC-LR inhibited the activity of protein phosphatase 2A (PP2A) in neurons, which is regarded as a molecular initiating event (MIE). In addition, the abnormalities in autophagy were observed after MC-LR exposure. The hindered autophagosome-lysosome fusion and disrupted lysosomal function were key events (KEs) after MC-LR exposure, which contributed to proteostasis dysregulation, ultimately leading to Aβ abnormal degradation and learning deficits as adverse outcomes (AO) of neurotoxicity. This study provided novel information about MC-LR neurotoxicity and new insights into understanding the mechanisms underlying the environmental chemicals-induced neurodegeneration diseases, which has deep implications for public health.
Collapse
Affiliation(s)
- Yuhan Ma
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-Embryology, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Dihui Xu
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-Embryology, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Yibin Gan
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-Embryology, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Zining Chen
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-Embryology, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Yabing Chen
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-Embryology, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, 210093, China.
| | - Xiaodong Han
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-Embryology, Medical School, Nanjing University, Nanjing, Jiangsu, 210093, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, 210093, China.
| |
Collapse
|
147
|
Zhang Z, Xue P, Bendlin BB, Zetterberg H, De Felice F, Tan X, Benedict C. Melatonin: A potential nighttime guardian against Alzheimer's. Mol Psychiatry 2025; 30:237-250. [PMID: 39128995 PMCID: PMC11649572 DOI: 10.1038/s41380-024-02691-6] [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: 02/28/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/13/2024]
Abstract
In the context of the escalating global health challenge posed by Alzheimer's disease (AD), this comprehensive review considers the potential of melatonin in both preventive and therapeutic capacities. As a naturally occurring hormone and robust antioxidant, accumulating evidence suggests melatonin is a compelling candidate to consider in the context of AD-related pathologies. The review considers several mechanisms, including potential effects on amyloid-beta and pathologic tau burden, antioxidant defense, immune modulation, and regulation of circadian rhythms. Despite its promise, several gaps need to be addressed prior to clinical translation. These include conducting additional randomized clinical trials in patients with or at risk for AD dementia, determining optimal dosage and timing, and further determining potential side effects, particularly of long-term use. This review consolidates existing knowledge, identifies gaps, and suggests directions for future research to better understand the potential of melatonin for neuroprotection and disease mitigation within the landscape of AD.
Collapse
Affiliation(s)
- Zefan Zhang
- Department of Big Data in Health Science, Zhejiang University School of Public Health and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China
| | - Pei Xue
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Barbara B Bendlin
- School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
- Wisconsin Alzheimer's Disease Research Center, Madison, WI, USA
- Wisconsin Alzheimer's Institute, Madison, WI, USA
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin, University of Wisconsin-Madison, Madison, WI, USA
| | - Fernanda De Felice
- Centre for Neurosciences Studies, Departments of Biomedical and Molecular Sciences, and Psychiatry, Queen's University, Kingston, ON, K7L 3N6, Canada
- D'Or Institute for Research and Education, Rio de Janeiro RJ, 22281-100, Brazil
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, 21941-902, Rio de Janeiro RJ, Brazil
| | - Xiao Tan
- Department of Big Data in Health Science, Zhejiang University School of Public Health and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China.
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Christian Benedict
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
148
|
Saribas AS, Jensen LE, Safak M. Recent advances in discovery and functional analysis of the small proteins and microRNA expressed by polyomaviruses. Virology 2025; 602:110310. [PMID: 39612622 DOI: 10.1016/j.virol.2024.110310] [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: 10/03/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 12/01/2024]
Abstract
The polyomavirus family consists of a highly diverse group of small DNA viruses isolated from various species, including humans. Some family members have been used as model systems to understand the fundamentals of modern biology. After the discovery of the first two human polyomaviruses (JC virus and BK virus) during the early 1970s, their current number reached 14 today. Some family members cause considerably severe human diseases, including polyomavirus-associated nephropathy (PVAN), progressive multifocal leukoencephalopathy (PML), trichodysplasia spinulosa (TS) and Merkel cell carcinoma (MCC). Polyomaviruses encode universal regulatory and structural proteins, but some members express additional virus-specific proteins and microRNA, which significantly contribute to the viral biology, cell transformation, and perhaps progression of the disease that they are associated with. In the current review, we summarized the recent advances in discovery, and functional and structural analysis of those viral proteins and microRNA.
Collapse
Affiliation(s)
- A Sami Saribas
- Lewis Katz School of Medicine at Temple University, Department of Microbiology, Immunology and Inflammation Center for Neurovirology and Gene Editing, 3500 N. Broad Street, Philadelphia, PA, 19140, USA.
| | - Liselotte E Jensen
- Lewis Katz School of Medicine at Temple University, Department of Microbiology, Immunology and Inflammation, Center for Inflammation and Lung Research, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Mahmut Safak
- Lewis Katz School of Medicine at Temple University, Department of Microbiology, Immunology and Inflammation Center for Neurovirology and Gene Editing, 3500 N. Broad Street, Philadelphia, PA, 19140, USA.
| |
Collapse
|
149
|
Anton PE, Maphis NM, Linsenbardt DN, Coleman LG. Excessive Alcohol Use as a Risk Factor for Alzheimer's Disease: Epidemiological and Preclinical Evidence. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1473:211-242. [PMID: 40128481 DOI: 10.1007/978-3-031-81908-7_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Alcohol use has recently emerged as a modifiable risk factor for Alzheimer's disease (AD). However, the neurobiological mechanisms by which alcohol interacts with AD pathogenesis remain poorly understood. In this chapter, we review the epidemiological and preclinical support for the interaction between alcohol use and AD. We hypothesize that alcohol use increases the rate of accumulation of specific AD-relevant pathologies during the prodromal phase and exacerbates dementia onset and progression. We find that alcohol consumption rates are increasing in adolescence, middle age, and aging populations. In tandem, rates of AD are also on the rise, potentially as a result of this increased alcohol use throughout the lifespan. We then review the biological processes in common between alcohol use disorder and AD as a means to uncover potential mechanisms by which they interact; these include oxidative stress, neuroimmune function, metabolism, pathogenic tauopathy development and spread, and neuronal excitatory/inhibitory balance (EIB). Finally, we provide some forward-thinking suggestions we believe this field should consider. In particular, the inclusion of alcohol use assessments in longitudinal studies of AD and more preclinical studies on alcohol's impacts using better animal models of late-onset Alzheimer's disease (LOAD).
Collapse
Affiliation(s)
- Paige E Anton
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Nicole M Maphis
- Department of Neurosciences and New Mexico Alcohol Research Center, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - David N Linsenbardt
- Department of Neurosciences and New Mexico Alcohol Research Center, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Leon G Coleman
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA.
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
150
|
Samal M, Srivastava V, Khan M, Insaf A, Penumallu NR, Alam A, Parveen B, Ansari SH, Ahmad S. Therapeutic Potential of Polyphenols in Cellular Reversal of Patho-Mechanisms of Alzheimer's Disease Using In Vitro and In Vivo Models: A Comprehensive Review. Phytother Res 2025; 39:25-50. [PMID: 39496498 DOI: 10.1002/ptr.8344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 07/28/2024] [Accepted: 08/31/2024] [Indexed: 11/06/2024]
Abstract
Alzheimer's disease (AD) is considered one of the most common neurological conditions associated with memory and cognitive impairment and mainly affects people aged 65 or above. Even with tremendous progress in modern neuroscience, a permanent remedy or cure for this crippling disease is still unattainable. Polyphenols are a group of naturally occurring potent compounds that can modulate the neurodegenerative processes typical of AD. The present comprehensive study has been conducted to find out the preclinical and clinical potential of polyphenols and elucidate their possible mechanisms in managing AD. Additionally, we have reviewed different clinical studies investigating polyphenols as single compounds or cotherapies, including those currently recruiting, completed, terminated, withdrawn, or suspended in AD treatment. Natural polyphenols were systematically screened and identified through electronic databases including Google Scholar, PubMed, and Scopus based on in vitro cell line studies and preclinical data demonstrating their potential for neuroprotection. A total of 63 significant polyphenols were identified. A multimechanistic pathway for polyphenol's mode of action has been proposed in the study. Out of 63, four potent polyphenols have been identified as promising potential candidates, based on their reported clinical efficacy. Polyphenols hold tremendous scope for the development of a future drug molecule as a phytopharmaceutical that may be incorporated as an adjuvant to the therapeutic regime. However, more high-quality studies with novel delivery methods and combinatorial approaches are required to overcome obstacles such as bioavailability and blood-brain barrier crossing to underscore the therapeutic potential of these compounds in AD management.
Collapse
Affiliation(s)
- Monalisha Samal
- Centre of Excellence in Unani Medicine, Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Varsha Srivastava
- Centre of Excellence in Unani Medicine, Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Muzayyana Khan
- Centre of Excellence in Unani Medicine, Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Areeba Insaf
- Centre of Excellence in Unani Medicine, Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Naveen Reddy Penumallu
- Centre of Excellence in Unani Medicine, Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Aftab Alam
- Centre of Excellence in Unani Medicine, Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Bushra Parveen
- Centre of Excellence in Unani Medicine, Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Shahid Hussain Ansari
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Sayeed Ahmad
- Centre of Excellence in Unani Medicine, Bioactive Natural Product Laboratory, Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| |
Collapse
|