1
|
Arayici ME, Kose A. Prevalence of Alzheimer's Disease and Cardiometabolic Multimorbidity in Older Adults Aged 60 and above in a Large-Scale Representative Sample in Türkiye: A Nationwide Population-Based Cross-Sectional Study. J Epidemiol Glob Health 2025; 15:86. [PMID: 40522427 DOI: 10.1007/s44197-025-00435-5] [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: 04/07/2025] [Accepted: 06/09/2025] [Indexed: 06/19/2025] Open
Abstract
BACKGROUND It is a well-established fact that Alzheimer's Disease (AD) is a growing public health challenge worldwide, with increasing prevalence among older adults. Cardiometabolic multimorbidity (CMM) and various sociodemographic and lifestyle factors are acknowledged to influence AD risk. This study aimed to estimate the prevalence of AD and investigate its association with CMM and related sociodemographic and behavioral factors in older adults aged 60 years and above in Türkiye. METHOD This study was conducted using secondary data obtained from the Türkiye Health Survey, provided by the Turkish Statistical Institute (TÜİK). Data were derived from 4630 individuals (weighted sample of 11,331,239) aged 60 years and older. AD prevalence, CMM (hypertension, coronary heart disease, stroke, diabetes, hyperlipidemia), and relevant covariates were evaluated. Multivariable binary logistic regression models were used to identify independent predictors of AD. RESULTS The overall prevalence of AD was 3.8% (95% CI: 3.6-4.9%), slightly higher in females (4.1%, 95% CI: 3.8-5.7%) than males (3.5%, 95% CI: 2.8-4.7%). AD was more common among individuals who were single/divorced (6.4%), had lower education (4.1%), presented with CMM (5.9%), or led sedentary lifestyles (4.9%). In the model adjusted for age, sex, educational status, and marital status, the presence of CMM was significantly associated with increased odds of AD (aOR = 2.081; 95% CI: 1.522-2.844). In the further adjusted model for BMI, tobacco use, alcohol use, and activity during the day, CMM remained a significant predictor of AD (aOR = 1.975, 95% CI: 1.446-2.698). CONCLUSIONS This large-scale study highlights a substantial burden of AD among older adults in Türkiye, strongly linked to CMM and modifiable social and behavioral factors. The findings of this study highlight the importance of integrated prevention strategies targeting vascular health, education, social support, and physical activity to reduce AD risk.
Collapse
Affiliation(s)
- Mehmet Emin Arayici
- Department of Public Health, Faculty of Medicine, Dokuz Eylül University, 15 July Medicine and Art Campus, Inciralti-Balcova 35340, Izmir, Turkey.
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Dokuz Eylül University, Izmir, Turkey.
| | - Ali Kose
- Department of Public Health, Institute of Health Sciences, Dokuz Eylül University, Izmir, Turkey
| |
Collapse
|
2
|
Meyer-Acosta KK, Diaz-Guerra E, Varma P, Aruk A, Mirsadeghi S, Muniz-Perez A, Rafati Y, Hosseini A, Nieto-Estevez V, Giugliano M, Navara C, Hsieh J. APOE4 impacts cortical neurodevelopment and alters network formation in human brain organoids. Stem Cell Reports 2025:102537. [PMID: 40541173 DOI: 10.1016/j.stemcr.2025.102537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 05/20/2025] [Accepted: 05/21/2025] [Indexed: 06/22/2025] Open
Abstract
Apolipoprotein E4 (APOE4) is the leading genetic risk factor for Alzheimer's disease. While most studies examine the role of APOE4 in aging, APOE4 causes persistent changes in brain structure as early as infancy and is associated with altered functional connectivity that extends beyond adolescence. Here, we used human induced pluripotent stem cell-derived cortical and ganglionic eminence organoids (COs and GEOs) to examine APOE4's influence during the development of cortical excitatory and inhibitory neurons. We show that APOE4 reduces cortical neurons and increases glia by promoting gliogenic transcriptional programs. In contrast, APOE4 increases proliferation and differentiation of GABAergic progenitors resulting in early and persistent increases in GABAergic neurons. Multi-electrode array recordings in assembloids revealed that APOE4 disrupts neural network function resulting in heightened excitability and synchronicity. Together, our data provide new insights on how APOE4 influences cortical neurodevelopmental processes and the establishment of functional networks.
Collapse
Affiliation(s)
- Karina K Meyer-Acosta
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA; Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Eva Diaz-Guerra
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA; Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Parul Varma
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA; Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Adyasha Aruk
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA; Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Sara Mirsadeghi
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA; Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Aranis Muniz-Perez
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA; Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Yousef Rafati
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA; Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Ali Hosseini
- International School of Advanced Studies, Neuroscience Area, V. Bonomea 265, 34136 Trieste, Italy
| | - Vanesa Nieto-Estevez
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA; Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Michele Giugliano
- Department of Biomedical, Metabolic & Neural Sciences, University of Modena and Reggio Emilia, V. Campi 287, 41125 Modena, Italy
| | - Christopher Navara
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA; Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA; Stem Cell Core, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Jenny Hsieh
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA; Brain Health Consortium, The University of Texas at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
3
|
Bigio B, Lima-Filho RAS, Barnhill O, Sudo FK, Drummond C, Assunção N, Vanderborght B, Beasley J, Young S, Korman A, Jones DR, Sultzer DL, Ferreira ST, Mattos P, Head E, Tovar-Moll F, De Felice FG, Lourenco MV, Nasca C. Sex differences in mitochondrial free-carnitine levels in subjects at-risk and with Alzheimer's disease in two independent study cohorts. Mol Psychiatry 2025; 30:2573-2583. [PMID: 39774493 DOI: 10.1038/s41380-024-02862-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 11/14/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025]
Abstract
A major challenge in the development of more effective therapeutic strategies for Alzheimer's disease (AD) is the identification of molecular mechanisms linked to specific pathophysiological features of the disease. Importantly AD has a two-fold higher incidence in women than men and a protracted prodromal phase characterized by amnestic mild-cognitive impairment (aMCI) suggesting that biological processes occurring early can initiate vulnerability to AD. Here, we used a sample of 125 subjects from two independent study cohorts to determine the levels in plasma (the most accessible specimen) of two essential mitochondrial markers acetyl-L-carnitine (LAC) and its derivative free-carnitine motivated by a mechanistic model in rodents in which targeting mitochondrial metabolism of LAC leads to the amelioration of cognitive function and boosts epigenetic mechanisms of gene expression. We report a sex-specific deficiency in free-carnitine levels in women with aMCI and early-AD compared to cognitively healthy controls; no change was observed in men. We also replicated the prior finding of decreased LAC levels in both women and men with AD, supporting the robustness of the study samples assayed in our new study. The magnitude of the sex-specific free-carnitine deficiency reflected the severity of cognitive dysfunction and held in two study cohorts. Furthermore, patients with the lower free-carnitine levels showed higher β-amyloid(Aβ) accumulation and t-Tau levels assayed in cerebrospinal fluid (CSF). Computational analyses showed that the mitochondrial markers assayed in plasma are at least as accurate as CSF measures to classify disease status. Together with the mechanistic platform in rodents, these translational findings lay the groundwork to create preventive individualized treatments targeting sex-specific changes in mitochondrial metabolism that may be subtle to early cognitive dysfunction of AD risk.
Collapse
Affiliation(s)
- Benedetta Bigio
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, USA
| | | | - Olivia Barnhill
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA
| | - Felipe K Sudo
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, RJ, Brazil
| | - Claudia Drummond
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, RJ, Brazil
- Department of Speech and Hearing Pathology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Naima Assunção
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, RJ, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Bart Vanderborght
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, RJ, Brazil
| | - James Beasley
- Biochemical Genetics Laboratory, Duke University Health System, Durham, NC, USA
| | - Sarah Young
- Biochemical Genetics Laboratory, Duke University Health System, Durham, NC, USA
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Aryeh Korman
- Metabolomics Laboratory, NYU Grossman School of Medicine, New York, NY, USA
| | - Drew R Jones
- Metabolomics Laboratory, NYU Grossman School of Medicine, New York, NY, USA
| | - David L Sultzer
- Department of Psychiatry and Human Behavior, School of Medicine, and Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, Irvine, CA, USA
| | - Sergio T Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Rio de Janeiro, RJ, Brazil
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, RJ, Brazil
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Paulo Mattos
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, RJ, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Institute of Psychiatry, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Elizabeth Head
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California Irvine, Irvine, CA, USA
- Department of Pathology and Laboratory Medicine, Department of Neurology, University of California Irvine, Irvine, CA, USA
| | - Fernanda Tovar-Moll
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, RJ, Brazil
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Rio de Janeiro, RJ, Brazil
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, RJ, Brazil
- Centre for Neurosciences Studies, Departments of Biomedical and Molecular Sciences & Department of Psychiatry, Queen's University, Kingston, ON, Canada
| | - Mychael V Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Rio de Janeiro, RJ, Brazil.
| | - Carla Nasca
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA.
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, USA.
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA.
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
4
|
Paul JK, Malik A, Azmal M, Gulzar T, Afghan MTR, Talukder OF, Shahzadi S, Ghosh A. Advancing Alzheimer's Therapy: Computational strategies and treatment innovations. IBRO Neurosci Rep 2025; 18:270-282. [PMID: 39995567 PMCID: PMC11849200 DOI: 10.1016/j.ibneur.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 01/22/2025] [Accepted: 02/02/2025] [Indexed: 02/26/2025] Open
Abstract
Alzheimer's disease (AD) is a multifaceted neurodegenerative condition distinguished by the occurrence of memory impairment, cognitive deterioration, and neuronal impairment. Despite extensive research efforts, conventional treatment strategies primarily focus on symptom management, highlighting the need for innovative therapeutic approaches. This review explores the challenges of AD treatment and the integration of computational methodologies to advance therapeutic interventions. A comprehensive analysis of recent literature was conducted to elucidate the broad scope of Alzheimer's etiology and the limitations of conventional drug discovery approaches. Our findings underscore the critical role of computational models in elucidating disease mechanisms, identifying therapeutic targets, and expediting drug discovery. Through computational simulations, researchers can predict drug efficacy, optimize lead compounds, and facilitate personalized medicine approaches. Moreover, machine learning algorithms enhance early diagnosis and enable precision medicine strategies by analyzing multi-modal datasets. Case studies highlight the application of computational techniques in AD therapeutics, including the suppression of crucial proteins implicated in disease progression and the repurposing of existing drugs for AD management. Computational models elucidate the interplay between oxidative stress and neurodegeneration, offering insights into potential therapeutic interventions. Collaborative efforts between computational biologists, pharmacologists, and clinicians are essential to translate computational insights into clinically actionable interventions, ultimately improving patient outcomes and addressing the unmet medical needs of individuals affected by AD. Overall, integrating computational methodologies represents a promising paradigm shift in AD therapeutics, offering innovative solutions to overcome existing challenges and transform the landscape of AD treatment.
Collapse
Affiliation(s)
- Jibon Kumar Paul
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Abbeha Malik
- Department of Bioinformatics, Institute of Biochemistry, Biotechnology and Bioinformatics, The Islamia University of Bahawalpur, Pakistan
| | - Mahir Azmal
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Tooba Gulzar
- Department of Bioinformatics, Institute of Biochemistry, Biotechnology and Bioinformatics, The Islamia University of Bahawalpur, Pakistan
| | - Muhammad Talal Rahim Afghan
- Department of Bioinformatics, Institute of Biochemistry, Biotechnology and Bioinformatics, The Islamia University of Bahawalpur, Pakistan
| | - Omar Faruk Talukder
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Samar Shahzadi
- Department of Bioinformatics, Institute of Biochemistry, Biotechnology and Bioinformatics, The Islamia University of Bahawalpur, Pakistan
| | - Ajit Ghosh
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| |
Collapse
|
5
|
Alami M, Morvaridzadeh M, El Khayari A, Boumezough K, El Fatimy R, Khalil A, Fulop T, Berrougui H. Reducing Alzheimer's disease risk with SGLT2 inhibitors: From glycemic control to neuroprotection. Ageing Res Rev 2025; 108:102751. [PMID: 40204129 DOI: 10.1016/j.arr.2025.102751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/28/2025] [Accepted: 04/04/2025] [Indexed: 04/11/2025]
Abstract
Recent research has established a strong link between metabolic abnormalities and an increased risk of dementia. In parallel, there is growing epidemiological evidence supporting the neuroprotective effects of antidiabetic medications against cognitive impairments. Among these, sodium-glucose co-transporter (SGLT2) inhibitors have emerged as pharmacological candidates with promising potential in alleviating the burden of age-related diseases, particularly neurodegenerative diseases (NDD). SGLT2 inhibitor therapies are FDA-approved medications routinely prescribed to manage diabetes. This novel class was initially developed to address cardiovascular disorders and to reduce the risk of hypoglycemia associated with insulin-secretagogue agents. It subsequently attracted growing interest for its beneficial effects on central nervous system (CNS) disorders. However, the molecular mechanisms through which these glucose-lowering therapies mitigate cognitive decline and limit the progression of certain brain degenerative diseases remain largely unexplored. Consequently, the neuroscientific community needs further studies that gather, analyze, and critically discuss the available mechanistic evidence regarding the neuroprotective effects of SGLT2 inhibitors. This review aims to critically examine the most relevant published findings, both in vitro and in vivo, as well as human studies evaluating the impact of SGLT2 inhibitors exposure on Alzheimer's disease (AD). It seeks to integrate the current understanding of their beneficial effects at the molecular level and their role in addressing the pathophysiology and neuropathology of AD. These insights will help extend our knowledge of how SGLT2 inhibitor therapies are associated with reduced risk of dementia and thus shed light on the link between diabetes and AD.
Collapse
Affiliation(s)
- Mehdi Alami
- Sultan Moulay Sliman University, Polydisciplinary Faculty, Department of Biology, Beni Mellal, Morocco; University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service, Sherbrooke, QC, Canada
| | - Mojgan Morvaridzadeh
- University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service, Sherbrooke, QC, Canada
| | - Abdellatif El Khayari
- Faculty of Medical Sciences, UM6P Hospitals, Mohammed VI Polytechnic University, Ben-Guerir 43150, Morocco; Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Kaoutar Boumezough
- Sultan Moulay Sliman University, Polydisciplinary Faculty, Department of Biology, Beni Mellal, Morocco; University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service, Sherbrooke, QC, Canada
| | - Rachid El Fatimy
- Faculty of Medical Sciences, UM6P Hospitals, Mohammed VI Polytechnic University, Ben-Guerir 43150, Morocco
| | - Abdelouahed Khalil
- University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service, Sherbrooke, QC, Canada
| | - Tamas Fulop
- University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service, Sherbrooke, QC, Canada
| | - Hicham Berrougui
- Sultan Moulay Sliman University, Polydisciplinary Faculty, Department of Biology, Beni Mellal, Morocco; University of Sherbrooke, Faculty of Medicine and Health Sciences, Department of Medicine, Geriatrics Service, Sherbrooke, QC, Canada.
| |
Collapse
|
6
|
Yokomizo S, Maci M, Stafford AM, Miller MR, Perle SJ, Takahashi S, Brown-Harding H, Liang L, Lovely A, Algamal M, Gillani RL, Zwang TJ, Richardson D, Naegele JR, Vogt D, Kastanenka KV. Transplantation of GABAergic Interneuron Progenitors Restores Cortical Circuit Function in an Alzheimer's Disease Mouse Model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.05.31.656412. [PMID: 40501774 PMCID: PMC12154612 DOI: 10.1101/2025.05.31.656412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/28/2025]
Abstract
In addition to dementia, Alzheimer's patients suffer from sleep impairments and aberrations in sleep-dependent brain rhythms. Deficits in inhibitory GABAergic interneuron function disrupt one of those rhythms, slow oscillation in particular, and actively contribute to Alzheimer's progression. We tested the degree to which transplantation of healthy donor interneuron progenitors would restore slow oscillation rhythm in young APP mice. We harvested medial ganglionic eminence (MGE) progenitors from mouse embryos and transplanted them into host APP mutant cortices. 3D light-sheet and structured illumination microscopy revealed that transplanted MGE progenitors survived and matured into healthy interneurons. In vivo multiphoton calcium imaging and voltage-sensitive dye imaging showed functional integration and slow oscillation rescue in absence or presence of optogenetic stimulation. Our work provides proof-of-concept evidence that stem cell therapy may serve as a viable strategy to rescue functional impairments in cortical circuits of APP mice and potentially those of Alzheimer's patients.
Collapse
Affiliation(s)
- Shinya Yokomizo
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Megi Maci
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - April M. Stafford
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, MI, USA
| | - Morgan R. Miller
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Stephen James Perle
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Shusaku Takahashi
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Heather Brown-Harding
- Department of Molecular and Cellular Biology and Harvard Center for Biological Imaging, Harvard University, Cambridge, MA, USA
| | - Linda Liang
- Department of Molecular and Cellular Biology and Harvard Center for Biological Imaging, Harvard University, Cambridge, MA, USA
| | - Alex Lovely
- Department of Molecular and Cellular Biology and Harvard Center for Biological Imaging, Harvard University, Cambridge, MA, USA
| | - Moustafa Algamal
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Rebecca L. Gillani
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Theodore J. Zwang
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Douglas Richardson
- Department of Molecular and Cellular Biology and Harvard Center for Biological Imaging, Harvard University, Cambridge, MA, USA
| | - Janice R. Naegele
- Department of Biology, Program in Neuroscience and Behavior, Hall-Atwater Laboratory, Wesleyan University, Middletown, CT, USA
| | - Daniel Vogt
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, MI, USA
| | - Ksenia V. Kastanenka
- Department of Neurology, MassGeneral Institute of Neurodegenerative Diseases, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
7
|
Alzarea SI, Alsaidan OA, Alhassan HH, Alzarea AI, Alsahli TG, Alharbi M, Afzal M, Sadiq Mantargi MJ. Neuraminidase as a novel therapeutic management strategy for Alzheimer's disease: evidenced through molecular docking, molecular dynamic simulation and gene expression analysis. Front Chem 2025; 13:1574702. [PMID: 40520681 PMCID: PMC12163420 DOI: 10.3389/fchem.2025.1574702] [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: 02/11/2025] [Accepted: 05/01/2025] [Indexed: 06/18/2025] Open
Abstract
Introduction Neuraminidase in humans is studied to see how well repurposed oseltamivir works for treating Alzheimer's disease (AD) using methods like molecular docking, molecular dynamic (MD) simulation, and gene expression analysis. Gene enrichment analysis was also studied to understand the behaviour of neuraminidases in humans. Methods Molecular docking was done using oseltamivir and the neuraminidase proteins with the PyRx tool, and the results were analysed using BIOVIA Discovery Studio. MD simulation (50 ns) of the oseltamivir and neuraminidase complex was performed using GROMACS tools. The gene expression analysis and gene enrichment study were done using GEO2R, which showed the results as log FC and significant values. Enricher tool-based gene enrichment analysis was done to determine the gene behaviour related to the AD. Results The molecular docking showed a strong connection between oseltamivir and neuraminidase (-6.5 kcal/mol), acetylcholinesterase (-7.9 kcal/mol), CDKs (-6.5 kcal/mol), and GSKs (-6.6 kcal/mol), interacting with different amino acids in the protein sequences. MD simulations showed a strong interaction between the ligand and neuraminidase, with stable measurements indicating that both the protein and ligand remained consistent in size and energy, which is better explained through the results of MM_PBSA and MM_GBSA analysis of the complex, resulting in the ΔE_vdW, ΔE_elec, ΔG_polar, ΔG_nonpolar, ΔG_gas, (ΔE_vdW + ΔEEL), ΔG_solvation: (ΔG_polar + ΔG_nonpolar) and ΔG_bind: total energies suggesting the complex stayed stable in conditions similar to those resembling natural cell. The gene expression analysis expressed TUBB3 (formation of beta-tubulin), FABP3 (regulates alpha-synuclein uptake in dopaminergic neurons), and CALM1 (calcium signal transduction pathway) to be highly upregulated in the given conditions with kinase binding (p = 0.0006541) and protein phosphatase regulatory activity (p = 0.001357) were highly upregulated, implicating their importance in the AD. Discussion The study ends on a hopeful note for using oseltamivir to treat neurological diseases, but it suggests that future research should include a solid cell line study, an in vitro study, and a clinical study.
Collapse
Affiliation(s)
- Sami I. Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
- King Salman Centre for Disability Research, Riyadh, Saudi Arabia
| | - Omar Awad Alsaidan
- King Salman Centre for Disability Research, Riyadh, Saudi Arabia
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | - Hassan H. Alhassan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | - Abdulaziz Ibrahim Alzarea
- Department of Clinical Pharmacy, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | - Tariq G. Alsahli
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | | |
Collapse
|
8
|
Dolgacheva LP, Zinchenko VP, Nadeev AD, Goncharov NV. Serotonergic Regulation in Alzheimer's Disease. Int J Mol Sci 2025; 26:5218. [PMID: 40508026 PMCID: PMC12154332 DOI: 10.3390/ijms26115218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 05/18/2025] [Accepted: 05/27/2025] [Indexed: 06/16/2025] Open
Abstract
Serotonin (5-HT) is a neurotransmitter that also plays an important role in the regulation of vascular tone and angiogenesis. This review focuses on the involvement of the 5-HT system in pathological processes leading to the development of Alzheimer's disease (AD). There is evidence that damage or dysfunction of the 5-HT system contributes to the development of AD, and different subtypes of 5-HT receptors are a potential target for the treatment of AD. A link has been established between AD, depression, stress, and 5-HT deficiency in the brain. There are new data on the role of circadian rhythms in modulating stress, depression, and the 5-HT system; amyloid β (Aβ) plaque clearance; and AD progression. Circadian disruption inhibits Aβ plaque clearance and modulates AD progression. The properties and functions of 5-HT, its receptors, and serotonergic neurons are presented. Special attention is paid to the central role of 5-HT in brain development, including neurite outgrowth, regulation of somatic morphology, motility, synaptogenesis, control of dendritic spine shape and density, neuronal plasticity determining its role in network regeneration, and changes in innervation after brain damage. The results of different studies indicate that the interaction of amyloid β oligomers (AβO) with mitochondria is a sufficient trigger for AD-related neurodegeneration. The action of 5-HT leads to an improvement in mitochondrial quality and the restoration of brain areas after traumatic brain injury, chronic stress, or developmental disorders in AD. The role of a healthy lifestyle and drugs acting on serotonin receptors in the prevention and treatment of AD is discussed.
Collapse
Affiliation(s)
- Lyudmila P. Dolgacheva
- Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, Institute of Cell Biophysics of the Russian Academy of Sciences, Pushchino 142290, Russia (V.P.Z.)
| | - Valery P. Zinchenko
- Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, Institute of Cell Biophysics of the Russian Academy of Sciences, Pushchino 142290, Russia (V.P.Z.)
| | - Alexander D. Nadeev
- Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, Institute of Cell Biophysics of the Russian Academy of Sciences, Pushchino 142290, Russia (V.P.Z.)
| | - Nikolay V. Goncharov
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, Saint Petersburg 194223, Russia
| |
Collapse
|
9
|
Lin CH, Wang SH, Lane HY. Sodium benzoate, a D-amino acid oxidase inhibitor, improved short-term memory in patients with mild cognitive impairment in a randomized, double-blind, placebo-controlled clinical trial. Psychiatry Clin Neurosci 2025. [PMID: 40405827 DOI: 10.1111/pcn.13841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/15/2025] [Accepted: 05/04/2025] [Indexed: 05/24/2025]
Abstract
BACKGROUND Previous studies have found that sodium benzoate (the pivotal D-amino acid oxidase [DAO] inhibitor) improved cognitive function in patients with mild Alzheimer disease; however, its efficacy for mild cognitive impairment (MCI) (especially its core feature, impaired short-term memory) remains uncertain. The aim of this study was to evaluate the efficacy and safety of sodium benzoate in treating amnestic MCI (aMCI). METHODS This study was a randomized, double-blind, placebo-controlled clinical trial conducted in a major medical center in Taiwan. Eighty-two patients with aMCI were recruited for 24-week treatment of 250 to 1500 mg/day of sodium benzoate or placebo. Overall, cognitive function was measured by Alzheimer's Disease Assessment Scale-Cognitive Subscale (ADAS-cog), and short-term memory was evaluated by the 'recall of test instructions' item in the ADAS-cog. The generalized estimating equation was applied to compare the two groups in efficacy. RESULTS Compared with placebo, sodium benzoate therapy, displayed a trend, albeit statistically insignificant, in improving overall cognitive function (P = 0.082), and significantly improved short-term memory (P = 0.044). Both benzoate and placebo were well tolerated and benzoate therapy produced no additional side effect. CONCLUSIONS With the moderate sample size of the current study, treatment using sodium benzoate, a DAO inhibitor, showed promise in improving cognition, especially short-term memory, in patients with aMCI. Of note, while the ADAS-cog total score has been regarded as insensitive in measuring aMCI, its 'recall of test instructions' item may be a more sensitive and clinically feasible tool. Further larger studies are warranted to confirm the preliminary finding. TRIAL REGISTRATION ClinicalTrials.gov Identifier. NCT04736355.
Collapse
Affiliation(s)
- Chieh-Hsin Lin
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Shi-Heng Wang
- National Center for Geriatrics and Welfare Research, National Health Research Institutes, Miaoli, Taiwan
| | - Hsien-Yuan Lane
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Psychiatry & Brain Disease Research Center, China Medical University Hospital, Taichung, Taiwan
- Department of Psychology, College of Medical and Health Sciences, Asia University, Taichung, Taiwan
| |
Collapse
|
10
|
Catto F, Kirschenbaum D, Economides AE, Reuss AM, Trevisan C, Caredio D, Dadgar-Kiani E, Mirzet D, Frick L, Weber-Stadlbauer U, Litvinov S, Koumoutsakos P, Lee JH, Aguzzi A. Quantitative 3D histochemistry reveals region-specific amyloid-β reduction by the antidiabetic drug netoglitazone. PLoS One 2025; 20:e0309489. [PMID: 40327707 PMCID: PMC12054868 DOI: 10.1371/journal.pone.0309489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 03/29/2025] [Indexed: 05/08/2025] Open
Abstract
A hallmark of Alzheimer's disease (AD) is the extracellular aggregation of toxic amyloid-beta (Aβ) peptides in form of plaques. Here, we identify netoglitazone, an antidiabetic compound previously tested in humans, as an Aβ aggregation antagonist. Netoglitazone improved cognition and reduced microglia activity in a mouse model of AD. Using quantitative whole-brain three-dimensional histology (Q3D), we precisely identified brain regions where netoglitazone reduced the number and size of Aβ plaques. We demonstrate the utility of Q3D in preclinical drug evaluation for AD by providing a high-resolution brain-wide view of drug efficacy. Applying Q3D has the potential to improve pre-clinical drug evaluation by providing information that can help identify mechanisms leading to brain region-specific drug efficacy.
Collapse
Affiliation(s)
- Francesca Catto
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- IMAI MedTech GmbH, Zurich, Switzerland
| | - Daniel Kirschenbaum
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Athena E. Economides
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Anna Maria Reuss
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Chiara Trevisan
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Davide Caredio
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ehsan Dadgar-Kiani
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zürich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Delic Mirzet
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Lukas Frick
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ulrike Weber-Stadlbauer
- Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zürich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Sergey Litvinov
- Computational Science and Engineering Laboratory, School of Engineering and Applied Sciences, Harvard University, Cambridge, United States of America
| | - Petros Koumoutsakos
- Computational Science and Engineering Laboratory, School of Engineering and Applied Sciences, Harvard University, Cambridge, United States of America
| | - Jin Hyung Lee
- Department of Neurology and Neurological Sciences, Stanford University, California, United States of America
- Department of Bioengineering, Stanford University, Stanford, California, United States of America
- Department of Electrical Engineering, Stanford University, Stanford, California, United States of America
- Department of Neurosurgery, Stanford University, Stanford, California, United States of America
| | - Adriano Aguzzi
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
11
|
Qneibi M, Bdir S, Bdair M, Aldwaik SA, Heeh M, Sandouka D, Idais T. Exploring the role of AMPA receptor auxiliary proteins in synaptic functions and diseases. FEBS J 2025; 292:2433-2478. [PMID: 39394632 DOI: 10.1111/febs.17287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 08/21/2024] [Accepted: 09/20/2024] [Indexed: 10/13/2024]
Abstract
α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) ionotropic glutamate receptors (AMPARs) mediate rapid excitatory synaptic transmission in the mammalian brain, primarily driven by the neurotransmitter glutamate. The modulation of AMPAR activity, particularly calcium-permeable AMPARs (CP-AMPARs), is crucially influenced by various auxiliary subunits. These subunits are integral membrane proteins that bind to the receptor's core and modify its functional properties, including ion channel kinetics and receptor trafficking. This review comprehensively catalogs all known AMPAR auxiliary proteins, providing vital insights into the biochemical mechanisms governing synaptic modulation and the specific impact of CP-AMPARs compared to their calcium-impermeable AMPA receptor (CI-AMPARs). Understanding the complex interplay between AMPARs and their auxiliary subunits in different brain regions is essential for elucidating their roles in cognitive functions such as learning and memory. Importantly, alterations in these auxiliary proteins' expression, function or interactions have been implicated in various neurological disorders. Aberrant signaling through CP-AMPARs, in particular, is associated with severe synaptic dysfunctions across neurodevelopmental, neurodegenerative and psychiatric conditions. Targeting the distinct properties of AMPAR-auxiliary subunit complexes, especially those involving CP-AMPARs, could disclose new therapeutic strategies, potentially allowing for more precise interventions in treating complex neuronal disorders.
Collapse
Affiliation(s)
- Mohammad Qneibi
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Sosana Bdir
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Mohammad Bdair
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Samia Ammar Aldwaik
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | | | - Dana Sandouka
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Tala Idais
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| |
Collapse
|
12
|
Bartolini E, Di Crosta A, La Malva P, Marin A, Ceccato I, Prete G, Mammarella N, Di Domenico A, Palumbo R. Gamma oscillation modulation for cognitive impairment: A systematic review. J Alzheimers Dis 2025; 105:331-350. [PMID: 40151908 DOI: 10.1177/13872877251328698] [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: 03/29/2025]
Abstract
BackgroundGamma oscillation modulation has emerged as a potential non-invasive treatment to counteract cognitive impairment in Alzheimer's disease (AD) and mild cognitive impairment (MCI). Non-invasive brain stimulation techniques like transcranial alternating current stimulation (tACS), gamma sensory stimulation (GSS), and transcranial magnetic stimulation (TMS) show promise in supporting specific cognitive functions.ObjectiveTo review and evaluate the efficacy of gamma oscillation modulation techniques in benefiting cognitive functions among individuals with AD and MCI.MethodsA systematic review was conducted, analyzing studies from 2015 to 2023 across databases such as PubMed, Web of Science, and Scopus. Inclusion criteria focused on studies involving tACS, GSS, or TMS applied to older adults with MCI or AD. A total of 438 articles were screened, of which 10 met the eligibility criteria.ResultsFindings suggest that gamma tACS, especially targeting the precuneus and dorsolateral prefrontal cortex, benefits episodic memory and cognitive performance. GSS also showed potential in supporting memory and attention, while TMS exhibited inconsistent but promising results when applied to the angular gyrus. However, heterogeneity in study designs and small sample sizes limit the generalizability of these outcomes.ConclusionsGamma oscillation modulation offers potential cognitive benefits for patients with AD and MCI, particularly in memory support. Further studies with larger samples and well-designed protocols are needed to confirm its therapeutic efficacy and optimize intervention parameters.
Collapse
Affiliation(s)
- Emanuela Bartolini
- Department of Psychology, University "G. d'Annunzio" of Chieti-Pescara, Chieti, CH, Italy
| | - Adolfo Di Crosta
- Department of Psychology, University "G. d'Annunzio" of Chieti-Pescara, Chieti, CH, Italy
| | - Pasquale La Malva
- Department of Psychology, University "G. d'Annunzio" of Chieti-Pescara, Chieti, CH, Italy
| | - Anna Marin
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Center for Translational Cognitive Neuroscience, VA Boston Healthcare System, Boston, MA, USA
| | - Irene Ceccato
- Department of Psychology, University "G. d'Annunzio" of Chieti-Pescara, Chieti, CH, Italy
| | - Giulia Prete
- Department of Psychology, University "G. d'Annunzio" of Chieti-Pescara, Chieti, CH, Italy
| | - Nicola Mammarella
- Department of Psychology, University "G. d'Annunzio" of Chieti-Pescara, Chieti, CH, Italy
| | - Alberto Di Domenico
- Department of Psychology, University "G. d'Annunzio" of Chieti-Pescara, Chieti, CH, Italy
| | - Rocco Palumbo
- Department of Psychology, University "G. d'Annunzio" of Chieti-Pescara, Chieti, CH, Italy
| |
Collapse
|
13
|
Wen H, He Y, Tang Y, Zhu L, Tao Q, Jin B, Luo T, Peng Y, Wei Y, Lei J, Wang L, Wang F, Ling F, Gao Y, Han L. Altered immune response is associated with sex difference in vulnerability to Alzheimer's disease in human prefrontal cortex. Brain Pathol 2025; 35:e13318. [PMID: 39497354 PMCID: PMC11961208 DOI: 10.1111/bpa.13318] [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: 05/27/2024] [Accepted: 10/17/2024] [Indexed: 04/03/2025] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder with a higher risk incidence in females than in males, and there are also differences in AD pathophysiology between sexes. The role of sex in the pathogenesis of AD may be crucial, yet the cellular and molecular basis remains unclear. Here, we performed a comprehensive analysis using four public transcriptome datasets of AD patients and age-matched control individuals in prefrontal cortex, including bulk transcriptome (295 females and 402 males) and single-nucleus RNA sequencing (snRNA-seq) data (224 females and 219 males). We found that the transcriptomic profile in female control was similar to those in AD. To characterize the key features associated with both the pathogenesis of AD and sex difference, we identified a co-expressed gene module that positively correlated with AD, sex, and aging, and was also enriched with immune-associated pathways. Using snRNA-seq datasets, we found that microglia (MG), a resident immune cell in the brain, demonstrated substantial differences in several aspects between sexes, such as an elevated proportion of activated MG, altered transcriptomic profile and cell-cell interaction between MG and other brain cell types in female control. Additionally, genes upregulated in female MG, such as TLR2, MERTK, SPP1, SLA, ACSL1, and FKBP5, had high confidence to be identified as biomarkers to distinguish AD status, and these genes also interacted with some approved drugs for treatment of AD. These findings underscore the altered immune response in female is associated with sex difference in susceptibility to AD, and the necessity of considering sex factors when developing AD biomarkers and therapeutic strategies, providing a scientific basis for further in-depth studies on sex differences in AD.
Collapse
Affiliation(s)
- Huiying Wen
- BGI ResearchHangzhouChina
- School of Biology and Biological EngineeringSouth China University of TechnologyGuangzhouChina
- BGI ResearchShenzhenChina
| | - Youzhe He
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Yuanchun Tang
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
- School of Life SciencesZhengzhou UniversityZhengzhouChina
| | - Langjian Zhu
- BGI ResearchHangzhouChina
- School of Biology and Biological EngineeringSouth China University of TechnologyGuangzhouChina
- BGI ResearchShenzhenChina
| | - Quyuan Tao
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Bufan Jin
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Ting Luo
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
| | - Yujie Peng
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
| | - Yanrong Wei
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Junjie Lei
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Lifang Wang
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
| | - Fan Wang
- Department of Pathology of Sir Run Run Shaw Hospital, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang University School of MedicineZhejiangHangzhouChina
- Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang University School of MedicineZhejiangHangzhouChina
| | - Fei Ling
- School of Biology and Biological EngineeringSouth China University of TechnologyGuangzhouChina
| | - Yue Gao
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
- Department of Pathology of Sir Run Run Shaw Hospital, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang University School of MedicineZhejiangHangzhouChina
- Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical NeurobiologyZhejiang University School of MedicineZhejiangHangzhouChina
| | - Lei Han
- BGI ResearchHangzhouChina
- BGI ResearchShenzhenChina
| |
Collapse
|
14
|
Mukherjee AG, Mishra S, Gopalakrishnan AV, Kannampuzha S, Murali R, Wanjari UR, B S, Vellingiri B, Madhyastha H, Kanagavel D, Vijayan M. Unraveling the mystery of citrate transporters in Alzheimer's disease: An updated review. Ageing Res Rev 2025; 107:102726. [PMID: 40073978 DOI: 10.1016/j.arr.2025.102726] [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: 06/25/2024] [Revised: 12/26/2024] [Accepted: 03/05/2025] [Indexed: 03/14/2025]
Abstract
A key molecule in cellular metabolism, citrate is essential for lipid biosynthesis, energy production, and epigenetic control. The etiology of Alzheimer's disease (AD), a progressive neurodegenerative illness marked by memory loss and cognitive decline, may be linked to dysregulated citrate transport, according to recent research. Citrate transporters, which help citrate flow both inside and outside of cells, are becoming more and more recognized as possible participants in the molecular processes underlying AD. Citrate synthase (CS), a key enzyme in the tricarboxylic acid (TCA) cycle, supports mitochondrial function and neurotransmitter synthesis, particularly acetylcholine (ACh), essential for cognition. Changes in CS activity affect citrate availability, influencing energy metabolism and neurotransmitter production. Choline, a precursor for ACh, is crucial for neuronal function. Lipid metabolism, oxidative stress reactions, and mitochondrial function can all be affected by aberrant citrate transport, and these changes are linked to dementia. Furthermore, the two main pathogenic characteristics of AD, tau hyperphosphorylation and amyloid-beta (Aβ) aggregation, may be impacted by disturbances in citrate homeostasis. The goal of this review is to clarify the complex function of citrate transporters in AD and provide insight into how they contribute to the development and course of the illness. We aim to provide an in-depth idea of which particular transporters are dysregulated in AD and clarify the functional implications of these dysregulated transporters in brain cells. To reduce neurodegenerative processes and restore metabolic equilibrium, we have also discussed the therapeutic potential of regulating citrate transport. Gaining insight into the relationship between citrate transporters and the pathogenesis of AD may help identify new indicators for early detection and creative targets for treatment. This study offers hope for more potent ways to fight this debilitating illness and is a crucial step in understanding the metabolic foundations of AD.
Collapse
Affiliation(s)
- Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Shatakshi Mishra
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, VIT, Vellore 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India.
| | - Sandra Kannampuzha
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Reshma Murali
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Stany B
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, VIT, Vellore 632014, India
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational Research, Department of Zoology, School of Basic Sciences, Central University of Punjab (CUPB), Bathinda, Punjab 151401, India
| | - Harishkumar Madhyastha
- Department of Cardiovascular Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki 8891692, Japan
| | - Deepankumar Kanagavel
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, VIT, Vellore 632014, India
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
15
|
Banerjee A, Thekkekkara D, Manjula SN, Nair SP, Lalitha MS. Correlation of autophagy and Alzheimer's disease with special emphasis on the role of phosphodiesterase-4. 3 Biotech 2025; 15:139. [PMID: 40292249 PMCID: PMC12018668 DOI: 10.1007/s13205-025-04306-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/02/2025] [Indexed: 04/30/2025] Open
Abstract
Autophagy disruption is important in Alzheimer's disease (AD) as it prevents misfolded proteins from being removed, which leads to the accumulation of amyloid plaques and neurofibrillary tangles (NFTs). Restoring autophagy improves neuronal survival and cognitive function, according to experimental models. In AD models, mTOR inhibition and AMPK activation enhance synaptic plasticity and lessen learning deficits. Inhibitors of phosphodiesterase-4 (PDE4) improve cognition and reduce neuroinflammation via altering cyclic adenosine monophosphate (cAMP) transmission. Furthermore, autophagic-lysosomal clearance is encouraged by upregulating transcription factor EB (TFEB), which lessens the pathogenic damage linked to AD. These results point to autophagy modification as a promising therapeutic approach, with the mTOR, AMPK, cAMP, and TFEB pathways being possible targets for drugs. Though much evidence is based on animal studies, these findings provide valuable insights into autophagy's role in AD pathology, offering promising directions for future research and drug development.
Collapse
Affiliation(s)
- Aniruddha Banerjee
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - Dithu Thekkekkara
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - S. N. Manjula
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - Salini P. Nair
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - Mankala Sree Lalitha
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| |
Collapse
|
16
|
Kalra P, Grewal AK, Khan H, Singh TG. Unscrambling the cellular and molecular threads of Neuroplasticity: Insights into Alzheimer's disease pathogenesis. Neuroscience 2025; 571:74-88. [PMID: 39970983 DOI: 10.1016/j.neuroscience.2025.02.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/14/2025] [Accepted: 02/15/2025] [Indexed: 02/21/2025]
Abstract
Alzheimer's disease (AD) is predominantly the most recurring and devastating neurological condition among the elderly population, characterized by the accumulation of amyloid-β (Aβ) and phosphorylated tau proteins, and is accompanied by progressive decline of learning and memory. Due to its complex and multifactorial etiology, a wide variety of therapeutic interventions have been developed. Despite constant advancements in the field, effective treatments that ameliorate the severity of Alzheimer's symptoms or cease their progression are still insufficient. Mounting evidence suggests that synaptic dysfunction could be an essential component of AD pathogenesis as synapse signaling is impaired in the aging brain, which contributes to synaptic decline. Therefore, improving neuroplasticity such as synaptic plasticity or neurogenesis could be a promising therapeutic approach for alleviating the effects of AD. This article reviews the cellular and molecular threads of neuroplasticity as well as targets that restore neuronal survival and plasticity to provide functional recoveries, including receptors, downstream signaling pathways, ion channels, transporters, enzymes, and neurotrophic factors.
Collapse
Affiliation(s)
- Palak Kalra
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India; University School of Pharmaceutical Sciences, Rayat Bahra University, Mohali, Punjab 140103, India
| | - Amarjot Kaur Grewal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India.
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India.
| |
Collapse
|
17
|
Ren Y, Pieper AA, Cheng F. Utilization of precision medicine digital twins for drug discovery in Alzheimer's disease. Neurotherapeutics 2025; 22:e00553. [PMID: 39965994 PMCID: PMC12047495 DOI: 10.1016/j.neurot.2025.e00553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/11/2025] [Accepted: 02/06/2025] [Indexed: 02/20/2025] Open
Abstract
Alzheimer's disease (AD) presents significant challenges in drug discovery and development due to its complex and poorly understood pathology and etiology. Digital twins (DTs) are recently developed virtual real-time representations of physical entities that enable rapid assessment of the bidirectional interaction between the virtual and physical domains. With recent advances in artificial intelligence (AI) and the growing accumulation of multi-omics and clinical data, application of DTs in healthcare is gaining traction. Digital twin technology, in the form of multiscale virtual models of patients or organ systems, can track health status in real time with continuous feedback, thereby driving model updates that enhance clinical decision-making. Here, we posit an additional role for DTs in drug discovery, with particular utility for complex diseases like AD. In this review, we discuss salient challenges in AD drug development, including complex disease pathology and comorbidities, difficulty in early diagnosis, and the current high failure rate of clinical trials. We also review DTs and discuss potential applications for predicting AD progression, discovering biomarkers, identifying new drug targets and opportunities for drug repurposing, facilitating clinical trials, and advancing precision medicine. Despite significant hurdles in this area, such as integration and standardization of dynamic medical data and issues of data security and privacy, DTs represent a promising approach for revolutionizing drug discovery in AD.
Collapse
Affiliation(s)
- Yunxiao Ren
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Andrew A Pieper
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH 44106, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland 44106, OH, USA; Department of Neurosciences, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA; Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - Feixiong Cheng
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA.
| |
Collapse
|
18
|
Huang J, Lai L, Su Y, Chen J, Li P, Du B. Probiotic-fermented ginger-processed Gastrodia elata BI. Ameliorates AlCl 3-induced cognitive dysfunction in an Alzheimer's disease rat model by regulating the gut microbiota and CREB/BDNF pathway. Food Res Int 2025; 207:116087. [PMID: 40086974 DOI: 10.1016/j.foodres.2025.116087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/24/2025] [Accepted: 02/22/2025] [Indexed: 03/16/2025]
Abstract
Gastrodia elata BI., which is an edible plant, has been reported in previous studies to possess a strong capacity for alleviating the symptoms of Alzheimer's disease (AD). This study focuses on ginger-processed and fermented Gastrodia elata BI. (FGGE) to investigate its effects on behaviour, brain neuroregulation, and the gut microbiota in an AlCl3-induced AD rat model, and to explore the underlying mechanisms. Results indicate that FGGE significantly improved novel object recognition and the correct alternation rate in the Y-maze test for AD rats. In addition, FGGE alleviated brain oxidative stress and restored the anti-inflammatory response, cholinergic function, and tissue morphology in the hippocampus. Furthermore, FGGE activated the cAMP response element-binding protein/brain-derived neurotrophic factor signalling pathway, reversing neural network abnormalities and enhancing neural regulation. FGGE also promoted the proliferation of bacteria negatively associated with AD, such as Methanosphaera and Lactobacillus, thereby restoring gut microbiota balance. The mechanisms by which FGGE alleviates AD may involve the modulation of the gut-brain axis, ultimately mitigating AD symptoms. FGGE represents an innovative functional food with significant therapeutic potential and promising application prospects.
Collapse
Affiliation(s)
- Junyuan Huang
- College of Food Science, South China Agricultural University, 510642 Guangzhou, China
| | - Lanyu Lai
- College of Food Science, South China Agricultural University, 510642 Guangzhou, China
| | - Yilin Su
- College of Food Science, South China Agricultural University, 510642 Guangzhou, China
| | - Jianping Chen
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Pan Li
- College of Food Science, South China Agricultural University, 510642 Guangzhou, China
| | - Bing Du
- College of Food Science, South China Agricultural University, 510642 Guangzhou, China.
| |
Collapse
|
19
|
Feng X, Jiang BW, Zhai SN, Liu CX, Wu H, Zhu BQ, Wei MY, Wei J, Yang L, Chen LL. Circular RNA aptamers targeting neuroinflammation ameliorate Alzheimer disease phenotypes in mouse models. Nat Biotechnol 2025:10.1038/s41587-025-02624-w. [PMID: 40164764 DOI: 10.1038/s41587-025-02624-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 03/03/2025] [Indexed: 04/02/2025]
Abstract
Alzheimer disease (AD) therapy may benefit from optimized approaches to inhibit neuroinflammation. Small-molecule inhibitors of the proinflammatory molecule double-stranded RNA (dsRNA)-activated protein kinase R (PKR) have efficacy in AD models but their utility is compromised by adverse side effects. Here, we target PKR in two mouse models of AD using circular RNAs containing short double-stranded regions (ds-cRNAs), which are structurally similar to what we used previously to target PKR in psoriasis models. We show that the intrahippocampal injection of ds-cRNAs to neurons and microglia by adeno-associated virus (AAV) effectively dampens excessive PKR activity with minimal toxicity, accompanied by reduced neuroinflammation and amyloid-β plaques. We also deliver ds-cRNAs to the whole brain through intravenous injection of AAV-PHP.eB, which crosses the blood-brain barrier, resulting in neuroprotection and enhanced capability of spatial learning and memory in AD mouse models. The delivery of ds-cRNAs at different progressive stages of AD alleviates disease phenotypes, with therapeutic effects sustained for at least 6 months after a single administration.
Collapse
Affiliation(s)
- Xin Feng
- Key Laboratory of RNA Innovation, Science and Engineering, New Cornerstone Science Laboratory, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Bo-Wen Jiang
- Key Laboratory of RNA Innovation, Science and Engineering, New Cornerstone Science Laboratory, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Si-Nan Zhai
- Center for Molecular Medicine, Children's Hospital of Fudan University and Shanghai Key Laboratory of Medical Epigenetics, International Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Chu-Xiao Liu
- Key Laboratory of RNA Innovation, Science and Engineering, New Cornerstone Science Laboratory, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hao Wu
- Key Laboratory of RNA Innovation, Science and Engineering, New Cornerstone Science Laboratory, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Bang-Qi Zhu
- Key Laboratory of RNA Innovation, Science and Engineering, New Cornerstone Science Laboratory, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Meng-Yuan Wei
- Key Laboratory of RNA Innovation, Science and Engineering, New Cornerstone Science Laboratory, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jia Wei
- Center for Molecular Medicine, Children's Hospital of Fudan University and Shanghai Key Laboratory of Medical Epigenetics, International Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Li Yang
- Center for Molecular Medicine, Children's Hospital of Fudan University and Shanghai Key Laboratory of Medical Epigenetics, International Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Ling-Ling Chen
- Key Laboratory of RNA Innovation, Science and Engineering, New Cornerstone Science Laboratory, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
- School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
- Shanghai Academy of Natural Sciences (SANS), Shanghai, China.
| |
Collapse
|
20
|
A E F Cardinali C, Martins YA, C M Moraes R, Costa AP, Torrão AS. Benfotiamine Ameliorates Streptozotocin-Induced Alzheimer's Disease in Rats by Modulating Neuroinflammation, Oxidative Stress, and Microglia. Mol Neurobiol 2025:10.1007/s12035-025-04811-x. [PMID: 40038195 DOI: 10.1007/s12035-025-04811-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 02/25/2025] [Indexed: 03/06/2025]
Abstract
Alzheimer's disease (AD) is the most prevalent cause of dementia, characterized by progressive memory loss and cognitive decline. Recent evidence indicates that inflammation plays a central role in AD pathogenesis, with elevated inflammatory markers and risk genes linked to innate immune functions. Glial cell dysfunction, particularly in astrocytes and microglia, is crucial to the neuroinflammatory process, contributing to oxidative stress, synaptic dysfunction, neuronal death, and impaired neurogenesis. This study aimed to investigate the therapeutic effects of benfotiamine (BFT), a vitamin B1 analogue, on microglial morphology, inflammation, and oxidative stress parameters in a sporadic Alzheimer-like disease model induced by intracerebroventricular injection of streptozotocin (STZ). Supplementation with 150 mg/kg of BFT for 7 days significantly reduced inflammation in the hippocampus and provided protection against oxidative damage in the entorhinal cortex by activating the Nrf-2 pathway and enhancing the expression of antioxidant enzymes such as SOD1 and CAT. These findings suggest that BFT exerts neuroprotective effects in AD, particularly impacting glial cell function and redox homeostasis.
Collapse
Affiliation(s)
- Camila A E F Cardinali
- Departamento de Fisiologia E Biofisica, Universidade de Sao Paulo, Av Professor Lineu Prestes 2415, Sao Paulo, 05508-000, Brazil.
| | - Yandara A Martins
- Departamento de Fisiologia E Biofisica, Universidade de Sao Paulo, Av Professor Lineu Prestes 2415, Sao Paulo, 05508-000, Brazil
| | - Ruan C M Moraes
- Departamento de Fisiologia E Biofisica, Universidade de Sao Paulo, Av Professor Lineu Prestes 2415, Sao Paulo, 05508-000, Brazil
- Department of Psychiatry & Behavioral Neurobiology, The University of Alabama at Birmingham, Alabama, USA
| | - Andressa P Costa
- Departamento de Fisiologia E Biofisica, Universidade de Sao Paulo, Av Professor Lineu Prestes 2415, Sao Paulo, 05508-000, Brazil
| | - Andréa S Torrão
- Departamento de Fisiologia E Biofisica, Universidade de Sao Paulo, Av Professor Lineu Prestes 2415, Sao Paulo, 05508-000, Brazil
| |
Collapse
|
21
|
Al‐Thani NA, Stewart GS, Costello DA. The Role of the Urea Cycle in the Alzheimer's Disease Brain. J Neurochem 2025; 169:e70033. [PMID: 40022483 PMCID: PMC11871420 DOI: 10.1111/jnc.70033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 02/13/2025] [Accepted: 02/13/2025] [Indexed: 03/03/2025]
Abstract
Alzheimer's Disease (AD) is a neurodegenerative disorder classified as the leading form of dementia in the elderly. Classical hallmarks of AD pathology believed to cause AD include Amyloid-beta (Aβ) plaques as well as neurofibrillary tau tangles (NTT). However, research into these classical hallmarks has failed to account for a causative link or therapeutic success. More recently, metabolic hallmarks of AD pathology have become a popular avenue of research. Elevated urea and ammonia detected in cases of AD point towards a dysfunctional urea cycle involved in AD. This review covers the expansive body of literature surrounding the work of researchers deciphering the role of the urea cycle in AD pathology through the study of urea cycle enzymes, metabolites, and transporters in the AD brain. Urea cycle enzymes of interest in AD pathology include OTC, NOS isoforms, ARG1, ARG2, MAOB, and ODC, which all present as promising therapeutic targets. Urea metabolites indicated in AD pathology have varying concentrations across the regions of the brain and the different cell types (neurons, microglia, astrocytes). Finally, the role of UT-B as a clearance modulator presents this protein as a key target for research in the role of the urea cycle in the AD brain. In the future, these key enzymes, pathways, and proteins relating to the urea cycle in AD should be further investigated to better understand the cell-specific urea cycle profiles in the AD brain and uncover their therapeutic potential.
Collapse
Affiliation(s)
- Najlaa A. Al‐Thani
- UCD School of Biomolecular and Biomedical ScienceUniversity College DublinDublinIreland
- UCD Conway InstituteUniversity College DublinDublinIreland
| | - Gavin S. Stewart
- UCD School of Biology and Environmental ScienceUniversity College DublinDublinIreland
| | - Derek A. Costello
- UCD School of Biomolecular and Biomedical ScienceUniversity College DublinDublinIreland
- UCD Conway InstituteUniversity College DublinDublinIreland
| |
Collapse
|
22
|
Ramsingh N, Lin HM, Ouyang Y, Ravona-Springer R, Livny A, Soleimani L, Bendlin BB, Matatov A, Niv T, Shamir T, Ganmore I, Heymann A, Sano M, Azuri J, Beeri MS. Brain Signatures of Very Early Cognitive Decline in Asymptomatic Middle-Aged Offspring of People With Alzheimer's Disease. Int J Geriatr Psychiatry 2025; 40:e70060. [PMID: 40050014 DOI: 10.1002/gps.70060] [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: 07/27/2024] [Revised: 12/17/2024] [Accepted: 02/11/2025] [Indexed: 05/13/2025]
Abstract
OBJECTIVE To assess the relationship of subtle preclinical cognitive changes with white matter microstructure and cortical volume in middle-aged adults at high AD risk due to a parental history. METHODS Participants (n = 278) were AD patients' offspring from the Israel Registry for Alzheimer's Prevention study. Cognitively unimpaired-decliners (CU-D) were based on a linear regression model. In a subsample with MRI (n = 220), we examined relationships of CU-D with white matter (WM) microstructure (fractional anisotropy [FA] and mean diffusivity [MD]) and cortical volume in brain regions commonly affected in AD. RESULTS CU-D participants had lower FA in the superior longitudinal fasciculus (SLF) (p = < 0.001) and higher MD in the SLF (p = < 0.001), and cingulum adjacent to the corpus callosum (p = < 0.001) and genu (p = 0.006) compared to cognitively unimpaired-stable (CU-S) participants. The groups did not differ in cortical brain volumes. CONCLUSIONS CU-D participants had poorer WM microstructure in brain tracts affected early in AD. Early interventions can target individuals that fit the CU-D criteria.
Collapse
Affiliation(s)
- Nadia Ramsingh
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Hung-Mo Lin
- Department of Anesthesiology and Yale Center for Analytical Sciences, Yale University, New Haven, Connecticut, USA
| | - Yuxia Ouyang
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ramit Ravona-Springer
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel HaShomer, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Psychiatry, Sheba Medical Center, Tel HaShomer, Israel
| | - Abigail Livny
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel HaShomer, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Division of Diagnostic Imaging, Sheba Medical Center, Tel HaShomer, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Laili Soleimani
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Barbara B Bendlin
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Adar Matatov
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel HaShomer, Israel
| | - Tal Niv
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel HaShomer, Israel
| | - Tamar Shamir
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel HaShomer, Israel
| | - Ithamar Ganmore
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel HaShomer, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Memory Clinic and Department of Neurology, Sheba Medical Center, Tel HaShomer, Israel
| | - Anthony Heymann
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Maccabi Health Services, Tel Aviv, Israel
- Meuhedat Healthcare, Tel Aviv, Israel
| | - Mary Sano
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- James J. Peters VA Medical Center, Bronx, New York, USA
| | | | - Michal Schnaider Beeri
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel HaShomer, Israel
- Krieger Klein Alzheimer's Research Center, Brain Health Institute, Rutgers University, New Brunswick, New Jersey, USA
| |
Collapse
|
23
|
Guo H, Yang R, Cheng W, Li Q, Du M. An Update of Salivary Biomarkers for the Diagnosis of Alzheimer's Disease. Int J Mol Sci 2025; 26:2059. [PMID: 40076682 PMCID: PMC11900270 DOI: 10.3390/ijms26052059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/23/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Alzheimer's disease (AD) is characterized by progressive cognition and behavior impairments. Diagnosing AD early is important for clinicians to slow down AD progression and preserve brain function. Biomarkers such as tau protein and amyloid-β peptide (Aβ) are used to aid diagnosis as clinical diagnosis often lags. Additionally, biomarkers can be used to monitor AD status and evaluate AD treatment. Clinicians detect these AD biomarkers in the brain using positron emission tomography/computed tomography or in the cerebrospinal fluid using a lumbar puncture. However, these methods are expensive and invasive. In contrast, saliva collection is simple, inexpensive, non-invasive, stress-free, and repeatable. Moreover, damage to the brain parenchyma can impact the oral cavity and some pathogenic molecules could travel back and forth from the brain to the mouth. This has prompted researchers to explore biomarkers in the saliva. Therefore, this study provides an overview of the main finding of salivary biomarkers for AD diagnosis. Based on these available studies, Aβ, tau, cholinesterase enzyme activity, lactoferrin, melatonin, cortisol, proteomics, metabolomics, exosomes, and the microbiome were changed in AD patients' saliva when compared to controls. However, well-designed studies are essential to confirm the reliability and validity of these biomarkers in diagnosing and monitoring AD.
Collapse
Affiliation(s)
| | | | | | | | - Minquan Du
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; (H.G.); (R.Y.); (W.C.); (Q.L.)
| |
Collapse
|
24
|
Naik RA, Mir MN, Malik IA, Bhardwaj R, Alshabrmi FM, Mahmoud MA, Alhomrani M, Alamri AS, Alsanie WF, Hjazi A, Ghatak T, Poeggeler B, Singh MP, Ts G, Singh SK. The Potential Mechanism and the Role of Antioxidants in Mitigating Oxidative Stress in Alzheimer's Disease. FRONT BIOSCI-LANDMRK 2025; 30:25551. [PMID: 40018917 DOI: 10.31083/fbl25551] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/30/2024] [Accepted: 08/23/2024] [Indexed: 03/01/2025]
Abstract
Alzheimer's disease (AD) is the most prevalent cause of dementia and a significant contributor to health issues and mortality among older individuals. This condition involves a progressive deterioration in cognitive function and the onset of dementia. Recent advancements suggest that the development of AD is more intricate than its underlying brain abnormalities alone. In addition, Alzheimer's disease, metabolic syndrome, and oxidative stress are all intricately linked to one another. Increased concentrations of circulating lipids and disturbances in glucose homeostasis contribute to the intensification of lipid oxidation, leading to a gradual depletion of the body's antioxidant defenses. This heightened oxidative metabolism adversely impacts cell integrity, resulting in neuronal damage. Pathways commonly acknowledged as contributors to AD pathogenesis include alterations in synaptic plasticity, disorganization of neurons, and cell death. Abnormal metabolism of some membrane proteins is thought to cause the creation of amyloid (Aβ) oligomers, which are extremely hazardous to neurotransmission pathways, especially those involving acetylcholine. The interaction between Aβ oligomers and these neurotransmitter systems is thought to induce cellular dysfunction, an imbalance in neurotransmitter signaling, and, ultimately, the manifestation of neurological symptoms. Antioxidants have a significant impact on human health since they may improve the aging process by combating free radicals. Neurodegenerative diseases are currently incurable; however, they may be effectively managed. An appealing alternative is the utilization of natural antioxidants, such as polyphenols, through diet or dietary supplements, which offer numerous advantages. Within this framework, we have extensively examined the importance of oxidative stress in the advancement of Alzheimer's disease, as well as the potential influence of antioxidants in mitigating its effects.
Collapse
Affiliation(s)
- Rayees Ahmad Naik
- Department of Zoology, Dr. Harisingh Gour Vishwavidyalaya Sagar, 470003 Sagar, Madhya Pradesh, India
| | - Mehak Naseer Mir
- NIMS Institute of Allied Medical Science, National Institute of Medical Sciences (NIMS), 303121 Jaipur, Rajasthan, India
| | - Ishfaq Ahmad Malik
- Department of Zoology, Bar. Ramrao Deshmukh Arts, Smt. Indiraji Kapadia Commerce & Nya. Krishnarao Deshmukh Science College, 444701 Amravati, Maharashtra, India
| | - Rima Bhardwaj
- Department of Chemistry Poona College, Savitribai Phule Pune University, 411007 Pune, Maharashtra, India
| | - Fahad M Alshabrmi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, 51452 Buraydah, Saudi Arabia
| | - Mahmoud Abdulrahman Mahmoud
- Department of Family & Community Medicine, College of Medicine, Imam Muhammad Ibn Saud Islamic University, 13313 Riyadh, Saudi Arabia
| | - Majid Alhomrani
- Department of Clinical Laboratory Sciences, The Faculty of Applied Medical Sciences, Taif University, 21944 Taif, Saudi Arabia
- Centre of Biomedical Sciences Research (CBSR), Deanship of Scientific Research, Taif University, 21944 Taif, Saudi Arabia
| | - Abdulhakeem S Alamri
- Department of Clinical Laboratory Sciences, The Faculty of Applied Medical Sciences, Taif University, 21944 Taif, Saudi Arabia
- Centre of Biomedical Sciences Research (CBSR), Deanship of Scientific Research, Taif University, 21944 Taif, Saudi Arabia
| | - Walaa F Alsanie
- Department of Clinical Laboratory Sciences, The Faculty of Applied Medical Sciences, Taif University, 21944 Taif, Saudi Arabia
- Centre of Biomedical Sciences Research (CBSR), Deanship of Scientific Research, Taif University, 21944 Taif, Saudi Arabia
| | - Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, 11942 Al-Kharj, Saudi Arabia
| | - Tanmoy Ghatak
- Department of Emergency Medicine, Sanjay Gandhi Post Graduate Institute of Medical Sciences, 226014 Lucknow, Uttar Pradesh, India
| | - Burkhard Poeggeler
- Department of Physiology, Johann-Friedrich-Blumenbach-Institute for Zoology and Anthropology, Faculty of Biology Georg August University Göttingen, Göttingen and Goettingen Research Campus, D-38524 Sassenburg, Germany
| | - Mahendra P Singh
- Department of Zoology, Deen Dayal Upadhyaya Gorakhpur University, 273009 Gorakhpur, Uttar Pradesh, India
| | - Gopenath Ts
- Department of Biotechnology & Bioinformatics, JSS Academy of Higher Education & Research, 570015 Mysuru, Karnataka, India
| | - Sandeep Kumar Singh
- Indian Scientific Education and Technology Foundation, 226001 Lucknow, Uttar Pradesh, India
| |
Collapse
|
25
|
Fu J, Wang R, He J, Liu X, Wang X, Yao J, Liu Y, Ran C, Ye Q, He Y. Pathogenesis and therapeutic applications of microglia receptors in Alzheimer's disease. Front Immunol 2025; 16:1508023. [PMID: 40028337 PMCID: PMC11867950 DOI: 10.3389/fimmu.2025.1508023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Microglia, the resident immune cells of the central nervous system, continuously monitor the brain's microenvironment through their array of specific receptors. Once brain function is altered, microglia are recruited to specific sites to perform their immune functions, including phagocytosis of misfolded proteins, cellular debris, and apoptotic cells to maintain homeostasis. When toxic substances are overproduced, microglia are over-activated to produce large amounts of pro-inflammatory cytokines, which induce chronic inflammatory responses and lead to neurotoxicity. Additionally, microglia can also monitor and protect neuronal function through microglia-neuron crosstalk. Microglia receptors are important mediators for microglia to receive external stimuli, regulate the functional state of microglia, and transmit signals between cells. In this paper, we first review the role of microglia-expressed receptors in the pathogenesis and treatment of Alzheimer's disease; moreover, we emphasize the complexity of targeting microglia for therapeutic interventions in neurodegenerative disorders to inform the discovery of new biomarkers and the development of innovative therapeutics.
Collapse
Affiliation(s)
- Jiao Fu
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
| | - RuoXuan Wang
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
| | - JiHui He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
| | - XiaoJing Liu
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
| | - XinXin Wang
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
| | - JuMing Yao
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
| | - Ye Liu
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - ChongZhao Ran
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - QingSong Ye
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yan He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
- Department of Stomatology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
26
|
Cho Y, Lee J, Kim JS, Jeon Y, Han S, Cho H, Lee Y, Kim TK, Hong JM, Lee Y, Byun Y, Chae M, Park S, Palomera LF, Park SY, Kim H, Kim S, Kang S, Jee JG, An H, Yim JH, Kim SH, Jo DG. RA-PR058, a novel ramalin derivative, reduces BACE1 expression and phosphorylation of tau in Alzheimer's disease mouse models. Anim Cells Syst (Seoul) 2025; 29:122-134. [PMID: 39931645 PMCID: PMC11809180 DOI: 10.1080/19768354.2025.2459649] [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: 11/21/2024] [Revised: 01/19/2025] [Accepted: 01/22/2025] [Indexed: 02/13/2025] Open
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disorder characterized by cognitive decline, anxiety-like behavior, β-amyloid (Aβ) accumulation, and tau hyperphosphorylation. BACE1, the enzyme critical for Aβ production, has been a major therapeutic target; however, direct BACE1 inhibition has been associated with adverse side effects. This study investigates the therapeutic potential of RA-PR058, a novel ramalin derivative, as a multi-targeted modulator of AD-related pathologies. The effects of RA-PR058 were evaluated in vitro and in vivo. In vitro studies used SH-SY5Y cells under oxidative stress conditions to assess BACE1 expression, while in vivo effects were studied in 3xTg-AD mice following one month of oral RA-PR058 treatment. Behavioral assessments, biochemical analyses, transcriptomic profiling, and pharmacokinetic evaluations were performed to determine the efficacy of RA-PR058. RA-PR058 significantly reduced oxidative stress-induced BACE1 expression in vitro and decreased cortical BACE1 expression in 3xTg-AD mice. In vivo treatment alleviated anxiety-like behavior and reduced tau phosphorylation at disease-relevant sites (Ser202/Thr205, Thr231, and Ser396). Transcriptomic analysis revealed RA-PR058-mediated gene expression changes related to central nervous system development, response to hypoxia, and neuroactive ligand-receptor interactions, suggesting broader regulatory effects on AD-related pathways. Pharmacokinetic analysis demonstrated that RA-PR058 exhibits high metabolic stability, minimal cytochrome P450 interactions, and moderate blood-brain barrier penetration. RA-PR058 demonstrates potential as a multi-target AD therapeutic by reducing BACE1 expression, tau hyperphosphorylation, and anxiety-like behavior, coupled with favorable pharmacokinetics. Additional studies are needed to assess cognitive effects and clarify molecular mechanisms, but RA-PR058 may represent a promising advancement in addressing AD's complex pathology.
Collapse
Affiliation(s)
- Yongeun Cho
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jeongmi Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jun-Sik Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yeji Jeon
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sukmin Han
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Heewon Cho
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yeongyeong Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Tai Kyoung Kim
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon, Republic of Korea
| | - Ju-Mi Hong
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon, Republic of Korea
| | - Yujeong Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yujung Byun
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Minshik Chae
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sunyoung Park
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Leon F. Palomera
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sang Yoon Park
- Bio Research Dept., Ahngook Pharmaceutical, Gwacheon, Republic of Korea
| | - Hyunwook Kim
- Bio Research Dept., Ahngook Pharmaceutical, Gwacheon, Republic of Korea
| | - Soyeong Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, Republic of Korea
| | - Seongeun Kang
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, Republic of Korea
| | - Jun-Goo Jee
- College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea
| | - Hongchan An
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, Republic of Korea
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pocheon, Republic of Korea
| | - Joung Han Yim
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon, Republic of Korea
| | - Sung Hyun Kim
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul, Republic of Korea
- Department of Physiology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
- Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, Republic of Korea
- Biomedical Institute for Convergence, Sungkyunkwan University, Suwon, Republic of Korea
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
27
|
Grahl MVC, Hohl KS, Smaniotto T, Carlini CR. Microbial Trojan Horses: Virulence Factors as Key Players in Neurodegenerative Diseases. Molecules 2025; 30:687. [PMID: 39942791 PMCID: PMC11820544 DOI: 10.3390/molecules30030687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/01/2025] [Accepted: 02/01/2025] [Indexed: 02/16/2025] Open
Abstract
Changes in population demographics indicate that the elderly population will reach 2.1 billion worldwide by 2050. In parallel, there will be an increase in neurodegenerative diseases such as Alzheimer's and Parkinson's. This review explores dysbiosis occurring in these pathologies and how virulence factors contribute to the worsening or development of clinical conditions, and it summarizes existing and potential ways to combat microorganisms related to these diseases. Microbiota imbalances can contribute to the progression of neurodegenerative diseases by increasing intestinal permeability, exchanging information through innervation, and even acting as a Trojan horse affecting immune cells. The microorganisms of the microbiota produce virulence factors to protect themselves from host defenses, many of which contribute to neurodegenerative diseases. These virulence factors are expressed according to the genetic composition of each microorganism, leading to a wide range of factors to be considered. Among the main virulence factors are LPS, urease, curli proteins, amyloidogenic proteins, VacA, and CagA. These factors can also be packed into bacterial outer membrane vesicles, which transport proteins, RNA, and DNA, enabling distal communication that impacts various diseases, including Alzheimer's and Parkinson's.
Collapse
Affiliation(s)
- Matheus V. C. Grahl
- Graduate Program in Medicine and Health Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90619-900, RS, Brazil
| | - Kelvin Siqueira Hohl
- Graduate Program in Biochemistry, Institute of Health Basic Sciences, Federal University of Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil; (K.S.H.); (T.S.)
| | - Thiago Smaniotto
- Graduate Program in Biochemistry, Institute of Health Basic Sciences, Federal University of Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil; (K.S.H.); (T.S.)
| | - Célia R. Carlini
- Center of Biotechnology, Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
- Graduate Program of Biosciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050-170, RS, Brazil
| |
Collapse
|
28
|
Wright AL, Weible AP, Estes OB, Wehr M. Ketamine does not rescue plaque load or gap detection in the 5XFAD mouse model of Alzheimer's disease. Front Aging Neurosci 2025; 17:1505908. [PMID: 39963471 PMCID: PMC11830726 DOI: 10.3389/fnagi.2025.1505908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
Ketamine has received growing attention for its effects on neuroplasticity and neuroinflammation, and as a treatment for depression and other mental health disorders. Recent evidence suggests that early sensory and behavioral deficits in Alzheimer's disease could be caused by synaptic disruption that occurs before irreversible neuropathology. This raises the possibility that ketamine could slow down or prevent network disruption and the ensuing sensory and behavioral deficits in Alzheimer's. Here we tested this idea in the 5XFAD mouse model of Alzheimer's, using either an acute single injection of ketamine, or chronic daily injections over 15 weeks. We tested the effects of ketamine on both amyloid plaque load and on a behavioral auditory gap detection task that is an early Alzheimer's biomarker in both mice and humans. We found that ketamine had no effect on plaque load, nor any effect on gap detection, for either acute or chronic dosing. Chronic ketamine facilitated startle responses specifically in 5XFAD mice, but this could simply be related to experience-dependent effects on stress or habituation rather than any rescue effect of ketamine on Alzheimer's-related deficits. We did find robust correlations between gap detection deficits and plaque load in auditory cortex and in the caudal pontine reticular nucleus, demonstrating that the behavioral deficits seen in 5XFAD mice are directly related to amyloid accumulation in these brain regions, and confirming the validity of gap detection as an early biomarker of Alzheimer's. Ketamine, however, had no effect on the strength of these correlations. We conclude that ketamine has no beneficial effect on the development of behavioral gap detection deficits or plaque load in the 5XFAD Alzheimer's mouse model, following either an acute single dose or a chronic daily dose regimen.
Collapse
Affiliation(s)
| | | | | | - Michael Wehr
- Department of Psychology, Institute of Neuroscience, University of Oregon, Eugene, OR, United States
| |
Collapse
|
29
|
Esandi J, Renault P, Capilla-López MD, Blanch R, Edo Á, Ramirez-Gómez D, Bosch A, Almolda B, Saura CA, Giraldo J, Chillón M. HEBE: A novel chimeric chronokine for ameliorating memory deficits in Alzheimer's disease. Biomed Pharmacother 2025; 183:117815. [PMID: 39818099 DOI: 10.1016/j.biopha.2025.117815] [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: 08/09/2024] [Revised: 01/05/2025] [Accepted: 01/09/2025] [Indexed: 01/18/2025] Open
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder characterized by amyloid-β and Tau protein depositions, with treatments focusing on single proteins have shown limited success due to the complexity of pathways involved. This study explored the potential of chronokines -proteins that modulate aging-related processes- as an alternative therapeutic approach. Specifically, we focused on a novel pleiotropic chimeric protein named HEBE, combining s-KL, sTREM2 and TIMP2, guided by bioinformatic analyses to ensure the preservation of each protein's conformation, crucial for their functions. In vitro studies confirmed HEBE's stability and enzymatic activities, even suggesting it has different activities compared to the individual chronokines. In vivo experiments on APP/Tau mice revealed improved learning and memory functions with HEBE treatment, along with decreased levels of phosphorylated Tau and minor effects on amyloid-β levels. These findings suggest that HEBE is as a promising therapeutic candidate for ameliorating memory deficits and reducing pTau in an AD mouse model.
Collapse
Affiliation(s)
- Jon Esandi
- Institut de Neurociències (INc), Universitat Autònoma Barcelona, Bellaterra 08193, Spain; Vall d'Hebron Institut de Recerca (VHIR), Barcelona 08035, Spain.
| | - Pedro Renault
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Unitat de Bioestadística, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain; Unitat de Neurociència Translacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Bellaterra 08193, Spain.
| | - Maria Dolores Capilla-López
- Institut de Neurociències (INc), Universitat Autònoma Barcelona, Bellaterra 08193, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28029, Spain.
| | - Rebeca Blanch
- Institut de Neurociències (INc), Universitat Autònoma Barcelona, Bellaterra 08193, Spain; Vall d'Hebron Institut de Recerca (VHIR), Barcelona 08035, Spain.
| | - Ángel Edo
- Institut de Neurociències (INc), Universitat Autònoma Barcelona, Bellaterra 08193, Spain; Vall d'Hebron Institut de Recerca (VHIR), Barcelona 08035, Spain.
| | - David Ramirez-Gómez
- Institut de Neurociències (INc), Universitat Autònoma Barcelona, Bellaterra 08193, Spain; Vall d'Hebron Institut de Recerca (VHIR), Barcelona 08035, Spain.
| | - Assumpció Bosch
- Institut de Neurociències (INc), Universitat Autònoma Barcelona, Bellaterra 08193, Spain; Vall d'Hebron Institut de Recerca (VHIR), Barcelona 08035, Spain.
| | - Beatriz Almolda
- Institut de Neurociències (INc), Universitat Autònoma Barcelona, Bellaterra 08193, Spain.
| | - Carlos Alberto Saura
- Institut de Neurociències (INc), Universitat Autònoma Barcelona, Bellaterra 08193, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28029, Spain.
| | - Jesús Giraldo
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Unitat de Bioestadística, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain; Unitat de Neurociència Translacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Bellaterra 08193, Spain.
| | - Miguel Chillón
- Institut de Neurociències (INc), Universitat Autònoma Barcelona, Bellaterra 08193, Spain; Vall d'Hebron Institut de Recerca (VHIR), Barcelona 08035, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona 08010, Spain.
| |
Collapse
|
30
|
Park JJ, Rim YA, Sohn Y, Nam Y, Ju JH. Prospects of induced pluripotent stem cells in treating advancing Alzheimer's disease: A review. Histol Histopathol 2025; 40:157-170. [PMID: 38847077 DOI: 10.14670/hh-18-766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
The World Health Organization has identified Alzheimer's disease (AD), the leading cause of dementia globally, as a public health priority. However, the complex multifactorial pathology of AD means that its etiology remains incompletely understood. Despite being recognized a century ago, incomplete knowledge has hindered the development of effective treatments for AD. Recent scientific advancements, particularly in induced pluripotent stem cell (iPSC) technology, show great promise in elucidating the fundamental mechanisms of AD. iPSCs play a dual role in regenerating damaged cells for therapeutic purposes and creating disease models to understand AD pathology and aid in drug screening. Nevertheless, as an emerging field, iPSC technology requires further technological advancement to develop effective AD treatments in the future. Thus, this review summarizes recent advances in stem cell therapies, specifically iPSCs, aimed at understanding AD pathology and developing treatments.
Collapse
Affiliation(s)
- Juyoun Janis Park
- YiPSCELL Inc, Seocho-gu, Seoul, South Korea
- Johns Hopkins University, Baltimore, Maryland, USA
| | - Yeri Alice Rim
- YiPSCELL Inc, Seocho-gu, Seoul, South Korea
- CiSTEM Laboratory, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Yeowon Sohn
- Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, South Korea
| | - Yoojun Nam
- YiPSCELL Inc, Seocho-gu, Seoul, South Korea
- Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, South Korea.
| | - Ji Hyeon Ju
- YiPSCELL Inc, Seocho-gu, Seoul, South Korea
- CiSTEM Laboratory, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea.
- Department of Biomedicine and Health Sciences, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
31
|
Agavriloaei LM, Iliescu BF, Pintilie RM, Turliuc DM. Therapeutic Potential of Experimental Stereotactic Hippocampal Cell Transplant in the Management of Alzheimer's Disease. J Clin Med 2025; 14:891. [PMID: 39941562 PMCID: PMC11818268 DOI: 10.3390/jcm14030891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/21/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
Due to a continuous increase in life expectancy and the progress made in specialized healthcare, the incidence of Alzheimer's disease (AD) has dramatically increased to the point that it has become one of the main challenges of contemporary medicine. Despite a huge scientific and clinical effort, current treatments manage just a temporary alleviation of symptomatology but offer no cure. Modern trials involving cell transplantation in experimental animals require the involvement of neurosurgeons in the treatment protocol. CSF shunting, intraventricular infusions, or DBS for symptoms relief have been an integral part of the therapeutic arsenal from the very beginning. The development of stereotactic surgery has facilitated the experimental potential of cell transplantation in the hippocampus for Alzheimer's disease. We conducted a narrative review of the literature in the top three medical databases (PubMed, Science Direct, and Google Scholar) using the keywords "Alzheimer's disease", "hippocampus", and "transplant". After eliminating duplicates, 241 papers were selected and screened by title and abstract. Two reviewers independently analyzed the 88 papers and chose 32 experiments that involved stereotactic hippocampal transplantation of cells in experimental animals with AD. The stereotactic transplantation of cells such as mesenchymal stem cells (MSCs), neuronal stem cells (NSCs), induced pluripotent cells (iPSCs), astrocytes, and derivates from stem cells was analyzed. The experiments used either a chemically induced or transgenic AD model and observed the impact of the stereotactic transplantation with behavioral testing, MRS spectroscopy, and biochemical analysis. The stereotaxic method delivers minimal invasive treatment option by cell transplantation at the hippocampus. The results showed that amyloid deposits were lower after transplantation, showing a positive impact. Other impactful results involve proliferation of neurogenesis, downregulation of anti-inflammatory response, and increased neuronal plasticity. The increased precision with which the stereotaxic method manages to target deep structures of the brain and the results of the reviewed papers could represent an argument for future human trials. More studies are needed to confirm the viability of the transplanted cells and the long-term effects.
Collapse
Affiliation(s)
- Loredana Mariana Agavriloaei
- Department of Neurosurgery, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (L.M.A.)
| | - Bogdan Florin Iliescu
- Department of Neurosurgery, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (L.M.A.)
- Department of Neurosurgery, “Prof. Dr. N. Oblu” Emergency Clinical Hospital, 700309 Iasi, Romania
| | - Robert Mihai Pintilie
- Department of Neurosurgery, “Prof. Dr. N. Oblu” Emergency Clinical Hospital, 700309 Iasi, Romania
| | - Dana Mihaela Turliuc
- Department of Neurosurgery, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (L.M.A.)
- Department of Neurosurgery, “Prof. Dr. N. Oblu” Emergency Clinical Hospital, 700309 Iasi, Romania
| |
Collapse
|
32
|
Bianchi O, Willey M, Alvarado CX, Danek B, Khani M, Kuznetsov N, Dadu A, Shah S, Koretsky MJ, Makarious MB, Weller C, Levine KS, Kim S, Jarreau P, Vitale D, Marsan E, Iwaki H, Leonard H, Bandres-Ciga S, Singleton AB, Nalls MA, Mokhtari S, Khashabi D, Faghri F. CARDBiomedBench: A Benchmark for Evaluating Large Language Model Performance in Biomedical Research: A novel question-and-answer benchmark designed to assess Large Language Models' comprehension of biomedical research, piloted on Neurodegenerative Diseases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.15.633272. [PMID: 39868292 PMCID: PMC11760394 DOI: 10.1101/2025.01.15.633272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Backgrounds Biomedical research requires sophisticated understanding and reasoning across multiple specializations. While large language models (LLMs) show promise in scientific applications, their capability to safely and accurately support complex biomedical research remains uncertain. Methods We present CARDBiomedBench, a novel question-and-answer benchmark for evaluating LLMs in biomedical research. For our pilot implementation, we focus on neurodegenerative diseases (NDDs), a domain requiring integration of genetic, molecular, and clinical knowledge. The benchmark combines expert-annotated question-answer (Q/A) pairs with semi-automated data augmentation, drawing from authoritative public resources including drug development data, genome-wide association studies (GWAS), and Summary-data based Mendelian Randomization (SMR) analyses. We evaluated seven private and open-source LLMs across ten biological categories and nine reasoning skills, using novel metrics to assess both response quality and safety. Results Our benchmark comprises over 68,000 Q/A pairs, enabling robust evaluation of LLM performance. Current state-of-the-art models show significant limitations: models like Claude-3.5-Sonnet demonstrates excessive caution (Response Quality Rate: 25% [95% CI: 25% ± 1], Safety Rate: 76% ± 1), while others like ChatGPT-4o exhibits both poor accuracy and unsafe behavior (Response Quality Rate: 37% ± 1, Safety Rate: 31% ± 1). These findings reveal fundamental gaps in LLMs' ability to handle complex biomedical information. Conclusion CARDBiomedBench establishes a rigorous standard for assessing LLM capabilities in biomedical research. Our pilot evaluation in the NDD domain reveals critical limitations in current models' ability to safely and accurately process complex scientific information. Future iterations will expand to other biomedical domains, supporting the development of more reliable AI systems for accelerating scientific discovery.
Collapse
Affiliation(s)
- Owen Bianchi
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- DataTecnica, Washington, DC, 20812, USA
| | - Maya Willey
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- DataTecnica, Washington, DC, 20812, USA
| | - Chelsea X. Alvarado
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- DataTecnica, Washington, DC, 20812, USA
| | - Benjamin Danek
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- DataTecnica, Washington, DC, 20812, USA
| | - Marzieh Khani
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Nicole Kuznetsov
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- DataTecnica, Washington, DC, 20812, USA
| | - Anant Dadu
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- DataTecnica, Washington, DC, 20812, USA
| | - Syed Shah
- DataTecnica, Washington, DC, 20812, USA
| | - Mathew J. Koretsky
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- DataTecnica, Washington, DC, 20812, USA
| | - Mary B. Makarious
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- DataTecnica, Washington, DC, 20812, USA
| | - Cory Weller
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- DataTecnica, Washington, DC, 20812, USA
| | - Kristin S. Levine
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- DataTecnica, Washington, DC, 20812, USA
| | - Sungwon Kim
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- DataTecnica, Washington, DC, 20812, USA
- Department of Computer Science, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Paige Jarreau
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Dan Vitale
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- DataTecnica, Washington, DC, 20812, USA
| | - Elise Marsan
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Hirotaka Iwaki
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- DataTecnica, Washington, DC, 20812, USA
| | - Hampton Leonard
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- DataTecnica, Washington, DC, 20812, USA
| | - Sara Bandres-Ciga
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Andrew B Singleton
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mike A Nalls
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- DataTecnica, Washington, DC, 20812, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | - Daniel Khashabi
- DataTecnica, Washington, DC, 20812, USA
- Department of Computer Science, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Faraz Faghri
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
- DataTecnica, Washington, DC, 20812, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
33
|
Zhou Q, Wu XN, Luo WH, Huang QH, Feng LL, Wu Y, Zhang C. Discovery of Effective Inhibitors Against Phosphodiesterase 9, a Potential Therapeutic Target of Alzheimer's Disease with Antioxidant Capacities. Antioxidants (Basel) 2025; 14:123. [PMID: 40002310 PMCID: PMC11852235 DOI: 10.3390/antiox14020123] [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: 11/15/2024] [Revised: 12/22/2024] [Accepted: 01/08/2025] [Indexed: 02/27/2025] Open
Abstract
Alzheimer's disease (AD) is a widely recognized type of dementia that leads to progressive cognitive decline and memory loss, affecting a significant number of people and their families worldwide. Given the multifactorial nature of AD, multitarget-directed ligands (MTDLs) hold promise in developing effective drugs for AD. Phosphodiesterase-9 (PDE9) is emerging as a promising target for AD therapy. In this study, by combining a PDE9 inhibitor C33 with the antioxidant melatonin, we designed and discovered a series of pyrazolopyrimidinone derivatives that simultaneously inhibit PDE9 and possess antioxidant activities. Molecular docking, together with dynamics simulations, were applied to accelerate compound design and reduce synthetic work. Four out of the 14 compounds were validated as effective PDE9 inhibitors with comparable antioxidant activity. Notably, compounds 17b and 17d demonstrated IC50 values of 91 and 89 nM against PDE9, respectively, with good antioxidant activities (ORAC (Trolox) of 2.00 and 2.60). This work provides a new approach for designing MTDLs for the treatment of AD and offers insights for further structural modifications of PDE9 inhibitors with antioxidant capacities.
Collapse
Affiliation(s)
- Qian Zhou
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, China; (Q.Z.); (W.-H.L.)
| | - Xu-Nian Wu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; (X.-N.W.); (Q.-H.H.); (L.-L.F.)
| | - Wei-Hao Luo
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, China; (Q.Z.); (W.-H.L.)
| | - Qing-Hua Huang
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; (X.-N.W.); (Q.-H.H.); (L.-L.F.)
| | - Ling-Ling Feng
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; (X.-N.W.); (Q.-H.H.); (L.-L.F.)
| | - Yinuo Wu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; (X.-N.W.); (Q.-H.H.); (L.-L.F.)
| | - Chen Zhang
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan 528458, China; (Q.Z.); (W.-H.L.)
| |
Collapse
|
34
|
Deak T, Burzynski HE, Nunes PT, Day SM, Savage LM. Adolescent Alcohol and the Spectrum of Cognitive Dysfunction in Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1473:257-298. [PMID: 40128483 DOI: 10.1007/978-3-031-81908-7_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Among the many changes associated with aging, inflammation in the central nervous system (CNS) and throughout the body likely contributes to the constellation of health-related maladies associated with aging. Genetics, lifestyle factors, and environmental experiences shape the trajectory of aging-associated inflammation, including the developmental timing, frequency, and intensity of alcohol consumption. This chapter posits that neuroinflammatory processes form a critical link between alcohol exposure and the trajectory of healthy aging, at least in part through direct or indirect interactions with cholinergic circuits that are crucial to cognitive integrity. In this chapter, we begin with a discussion of how inflammation changes from early development through late aging; discuss the role of inflammation and alcohol in the emergence of mild cognitive impairment (MCI); elaborate on critical findings on the contribution of alcohol-related thiamine deficiency to the loss of cholinergic function and subsequent development of Wernicke-Korsakoff syndrome (WKS); and present emerging findings at the intersection of alcohol and Alzheimer's disease and related dementias (ADRD). In doing so, our analysis points toward inflammation-mediated compromise of basal forebrain cholinergic function as a key culprit in cognitive dysfunction associated with chronic alcohol exposure, effects that may be rescuable through either pharmacological or behavioral approaches. Furthermore, our chapter reveals an interesting dichotomy in the effects of alcohol on neuropathological markers of ADRD that depend upon both biological sex and genetic vulnerability.
Collapse
Affiliation(s)
- Terrence Deak
- Developmental Exposure Alcohol Research Center (DEARC), Behavioral Neuroscience Program, Department of Psychology, Binghamton University-State University of New York, Binghamton, NY, USA.
| | - Hannah E Burzynski
- Developmental Exposure Alcohol Research Center (DEARC), Behavioral Neuroscience Program, Department of Psychology, Binghamton University-State University of New York, Binghamton, NY, USA
| | - Polliana T Nunes
- Developmental Exposure Alcohol Research Center (DEARC), Behavioral Neuroscience Program, Department of Psychology, Binghamton University-State University of New York, Binghamton, NY, USA
| | - Stephen M Day
- Developmental Exposure Alcohol Research Center (DEARC), Behavioral Neuroscience Program, Department of Psychology, Binghamton University-State University of New York, Binghamton, NY, USA
| | - Lisa M Savage
- Developmental Exposure Alcohol Research Center (DEARC), Behavioral Neuroscience Program, Department of Psychology, Binghamton University-State University of New York, Binghamton, NY, USA
| |
Collapse
|
35
|
Parul, Singh A, Shukla S. Novel techniques for early diagnosis and monitoring of Alzheimer's disease. Expert Rev Neurother 2025; 25:29-42. [PMID: 39435792 DOI: 10.1080/14737175.2024.2415985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 10/09/2024] [Indexed: 10/23/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) is the most common neurodegenerative disorder, which is characterized by a progressive loss of cognitive functions. The high prevalence, chronicity, and multimorbidity are very common in AD, which significantly impair the quality of life and functioning of patients. Early detection and accurate diagnosis of Alzheimer's disease (AD) can stop the illness from progressing thereby postponing its symptoms. Therefore, for the early diagnosis and monitoring of AD, more sensitive, noninvasive, straightforward, and affordable screening tools are needed. AREAS COVERED This review summarizes the importance of early detection methods and novel techniques for Alzheimer's disease diagnosis that can be used by healthcare professionals. EXPERT OPINION Early diagnosis assists the patient and caregivers to understand the problem establishing reasonable goals and making future plans together. Early diagnosis techniques not only help in monitoring disease progression but also provide crucial information for the development of novel therapeutic targets. Researchers can plan to potentially alleviate symptoms or slow down the progression of Alzheimer's disease by identifying early molecular changes and targeting altered pathways.
Collapse
Affiliation(s)
- Parul
- Division of Neuroscience and Ageing biology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Animesh Singh
- Division of Neuroscience and Ageing biology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Shubha Shukla
- Division of Neuroscience and Ageing biology, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
36
|
Kumar U. Cannabinoids: Role in Neurological Diseases and Psychiatric Disorders. Int J Mol Sci 2024; 26:152. [PMID: 39796008 PMCID: PMC11720483 DOI: 10.3390/ijms26010152] [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: 11/19/2024] [Revised: 12/20/2024] [Accepted: 12/22/2024] [Indexed: 01/13/2025] Open
Abstract
An impact of legalization and decriminalization of marijuana is the gradual increase in the use of cannabis for recreational purposes, which poses a potential threat to society and healthcare systems worldwide. However, the discovery of receptor subtypes, endogenous endocannabinoids, and enzymes involved in synthesis and degradation, as well as pharmacological characterization of receptors, has led to exploration of the use of cannabis in multiple peripheral and central pathological conditions. The role of cannabis in the modulation of crucial events involving perturbed physiological functions and disease progression, including apoptosis, inflammation, oxidative stress, perturbed mitochondrial function, and the impaired immune system, indicates medicinal values. These events are involved in most neurological diseases and prompt the gradual progression of the disease. At present, several synthetic agonists and antagonists, in addition to more than 70 phytocannabinoids, are available with distinct efficacy as a therapeutic alternative in different pathological conditions. The present review aims to describe the use of cannabis in neurological diseases and psychiatric disorders.
Collapse
Affiliation(s)
- Ujendra Kumar
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
37
|
Yang F, Gao W, Wang J, Li X, Li H. Progress of Chinese Medicine in Regulating Microglial Polarization against Alzheimer's Disease. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:2255-2275. [PMID: 39721955 DOI: 10.1142/s0192415x24500873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
Alzheimer's disease (AD), the predominant form of dementia, is a neurodegenerative disorder of the central nervous system (CNS) characterized by a subtle onset and a spectrum of cognitive and functional declines. The clinical manifestation of AD encompasses memory deficits, cognitive deterioration, and behavioral disturbances, culminating in a severe impairment of daily living skills. Despite its high prevalence, accounting for 60-70% of all dementia cases, there remains an absence of curative therapeutics. Microglia (MG), the resident immune cells of the CNS, exhibit a bifurcated role in AD pathogenesis. Functioning in a neuroprotective capacity, MGs express scavenger receptors, facilitating the clearance of [Formula: see text]-amyloid protein (A[Formula: see text]) and cellular debris. Conversely, aberrant activation of MGs can lead to the secretion of pro-inflammatory cytokines, thereby propagating neuroinflammatory responses that are detrimental to neuronal integrity. The dynamics of MG activation and the ensuing neuroinflammation are pivotal in the evolution of AD. Chinese medicine (CM), a treasure trove of traditional Chinese cultural practices, has demonstrated significant potential in the therapeutic management of AD. Over the past triennium, CM has garnered considerable research attention for its multifaceted approaches to AD, including the regulation of MG polarization. This review synthesizes current knowledge on the origins, polarization dynamics, and mechanistic interplay of MG with AD pathology. It further explores the nexus between MG polarization and cardinal pathological hallmarks of AD, such as A[Formula: see text] plaque deposition, hyperphosphorylation of tau, synaptic plasticity impairments, neuroinflammation, and brain-gut-axis dysregulation. The review also encapsulates the therapeutic strategies of CM, which encompass monomers, formulae, and acupuncture. These strategies modulate MG polarization in the context of AD treatment, thereby providing a robust theoretical framework in which to conduct future investigative endeavors in both the clinical and preclinical realms.
Collapse
Affiliation(s)
- Fengge Yang
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P. R. China
| | - Wei Gao
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P. R. China
- Jiangsu College of Nursing Jiangsu, Huaian, Huaiyin 223001, P. R. China
| | - Junting Wang
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P. R. China
- The Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150001, P. R. China
| | - Xue Li
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P. R. China
| | - Honglin Li
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150040, P. R. China
- The Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150001, P. R. China
| |
Collapse
|
38
|
Wright SA, Lennon R, Greenhalgh AD. Basement membranes' role in immune cell recruitment to the central nervous system. J Inflamm (Lond) 2024; 21:53. [PMID: 39707430 DOI: 10.1186/s12950-024-00426-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 12/10/2024] [Indexed: 12/23/2024] Open
Abstract
Basement membranes form part of the extracellular matrix (ECM), which is the structural basis for all tissue. Basement membranes are cell-adherent sheets found between cells and vascular endothelia, including those of the central nervous system (CNS). There is exceptional regional specialisation of these structures, both in tissue organisation and regulation of tissue-specific cellular processes. Due to their location, basement membranes perform a key role in immune cell trafficking and therefore are important in inflammatory processes causing or resulting from CNS disease and injury. This review will describe basement membranes in detail, with special focus on the brain. We will cover how genetic changes drive brain pathology, describe basement membranes' role in immune cell recruitment and how they respond to various brain diseases. Understanding how basement membranes form the junction between the immune and central nervous systems will be a major advance in understanding brain disease.
Collapse
Affiliation(s)
- Shaun A Wright
- Lydia Becker Institute of Immunology and Inflammation, Division, Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Science, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Rachel Lennon
- Cell Matrix Biology & Regenerative Medicine and Wellcome Centre for Cell-Matrix Research, School of Biological Science, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Andrew D Greenhalgh
- Lydia Becker Institute of Immunology and Inflammation, Division, Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Science, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
- The University of Manchester, Oxford Road, Manchester, M13 9PT, UK.
| |
Collapse
|
39
|
Del Rosario Hernandez T, Joshi NR, Gore SV, Kreiling JA, Creton R. Combining supervised and unsupervised analyses to quantify behavioral phenotypes and validate therapeutic efficacy in a triple transgenic mouse model of Alzheimer's disease. Biomed Pharmacother 2024; 181:117718. [PMID: 39637754 PMCID: PMC11755788 DOI: 10.1016/j.biopha.2024.117718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/11/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024] Open
Abstract
Behavioral testing is an essential tool for evaluating cognitive function and dysfunction in preclinical research models. This is of special importance in the study of neurological disorders such as Alzheimer's disease. However, the reproducibility of classic behavioral assays is frequently compromised by interstudy variation, leading to ambiguous conclusions about the behavioral markers characterizing the disease. Here, we identify age- and genotype-driven differences between 3xTg-AD and non-transgenic control mice using a low-cost, highly customizable behavioral assay that requires little human intervention. Through behavioral phenotyping combining both supervised and unsupervised behavioral classification methods, we are able to validate the preventative effects of the immunosuppressant cyclosporine A in a rodent model of Alzheimer's disease, as well as the partially ameliorating effects of candidate drugs nebivolol and cabozantinib.
Collapse
Affiliation(s)
- Thais Del Rosario Hernandez
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI United States.
| | - Narendra R Joshi
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI United States
| | - Sayali V Gore
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI United States
| | - Jill A Kreiling
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI United States
| | - Robbert Creton
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI United States
| |
Collapse
|
40
|
Singh M, Ali H, Renuka Jyothi S, Kaur I, Kumar S, Sharma N, Siva Prasad GV, Pramanik A, Hassan Almalki W, Imran M. Tau proteins and senescent Cells: Targeting aging pathways in Alzheimer's disease. Brain Res 2024; 1844:149165. [PMID: 39155034 DOI: 10.1016/j.brainres.2024.149165] [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: 06/16/2024] [Revised: 08/02/2024] [Accepted: 08/12/2024] [Indexed: 08/20/2024]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disease characterized by abnormal accumulation of tau proteins and amyloid-β, leading to neuronal death and cognitive impairment. Recent studies have implicated aging pathways, including dysregulation of tau and cellular senescence in AD pathogenesis. In AD brains, tau protein, which normally stabilizes microtubules, becomes hyperphosphorylated and forms insoluble neurofibrillary tangles. These tau aggregates impair neuronal function and are propagated across the brain's neurocircuitry. Meanwhile, the number of senescent cells accumulating in the aging brain is rising, releasing a pro-inflammatory SASP responsible for neuroinflammation and neurodegeneration. This review explores potential therapeutic interventions for AD targeting tau protein and senescent cells, and tau -directed compounds, senolytics, eliminating senescent cells, and agents that modulate the SASP-senomodulators. Ultimately, a combined approach that incorporates tau-directed medications and targeted senescent cell-based therapies holds promise for reducing the harmful impact of AD's shared aging pathways.
Collapse
Affiliation(s)
- Mahaveer Singh
- School of Pharmacy and Technology Management, SVKMs NMIMS University, Shirpur campus, Maharastra India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - S Renuka Jyothi
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Irwanjot Kaur
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan-303012, India
| | - Sachin Kumar
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Naveen Sharma
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali 140307, Punjab, India
| | - G V Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Atreyi Pramanik
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| |
Collapse
|
41
|
Wang P, Okada-Rising S, Scultetus AH, Bailey ZS. The Relevance and Implications of Monoclonal Antibody Therapies on Traumatic Brain Injury Pathologies. Biomedicines 2024; 12:2698. [PMID: 39767605 PMCID: PMC11672875 DOI: 10.3390/biomedicines12122698] [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: 09/25/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
Traumatic brain injury (TBI) is a global public health concern. It remains one of the leading causes of morbidity and mortality. TBI pathology involves complex secondary injury cascades that are associated with cellular and molecular dysfunction, including oxidative stress, coagulopathy, neuroinflammation, neurodegeneration, neurotoxicity, and blood-brain barrier (BBB) dysfunction, among others. These pathological processes manifest as a diverse array of clinical impairments. They serve as targets for potential therapeutic intervention not only in TBI but also in other diseases. Monoclonal antibodies (mAbs) have been used as key therapeutic agents targeting these mechanisms for the treatment of diverse diseases, including neurological diseases such as Alzheimer's disease (AD). MAb therapies provide a tool to block disease pathways with target specificity that may be capable of mitigating the secondary injury cascades following TBI. This article reviews the pathophysiology of TBI and the molecular mechanisms of action of mAbs that target these shared pathological pathways in a wide range of diseases. Publicly available databases for various applications of mAb therapy were searched and further classified to assess relevance to TBI pathology and evaluate current stages of development. The authors intend for this review to highlight the potential impact of current mAb technology within pathological TBI processes.
Collapse
Affiliation(s)
- Ping Wang
- Brain Trauma Neuroprotection, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; (S.O.-R.); (A.H.S.); (Z.S.B.)
| | | | | | | |
Collapse
|
42
|
Al-Thani NA, Zinck D, Stewart GS, Costello DA. Modulation of Urea Transport Attenuates TLR2-Mediated Microglial Activation and Upregulates Microglial Metabolism In Vitro. Metabolites 2024; 14:634. [PMID: 39590870 PMCID: PMC11596256 DOI: 10.3390/metabo14110634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/06/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Alzheimer's disease (AD) is a neurodegenerative disorder traditionally characterised by the presence of amyloid beta (Aβ) plaques and neurofibrillary tau tangles in the brain. However, emerging research has highlighted additional metabolic hallmarks of AD pathology. These include the metabolic reprogramming of microglia in favour of glycolysis over oxidative phosphorylation. This shift is attributed to an 'M1'-like pro-inflammatory phenotype, which exacerbates neuroinflammation and contributes to neuronal damage. The urea cycle also presents as an altered metabolic pathway in AD, due to elevated urea levels and altered expression of urea cycle enzymes, metabolites, and transporters in the brain. However, to date, these changes remain largely unexplored. Methods: This study focuses on understanding the effects of extracellular urea and urea transporter-B (UT-B) inhibition on inflammatory changes in lipoteichoic acid (LTA)-stimulated BV2 microglia and on the viability of SH-SY5Y neuronal cells under oxidative stress and neurotoxic conditions. Results: In BV2 microglia, UT-B inhibition demonstrated a notable anti-inflammatory effect by reducing the formation of nitric oxide (NO) and the expression of tumour necrosis factor α (TNFα) and CCL2 in response to stimulation with the toll-like receptor (TLR)2 agonist, lipoteichoic acid (LTA). This was accompanied by a reduction in extracellular urea and upregulation of UT-B expression. The application of exogenous urea was also shown to mediate the inflammatory profile of BV2 cells in a similar manner but had only a modest impact on UT-B expression. While exposure to LTA alone did not alter the microglial metabolic profile, inhibition of UT-B upregulated the expression of genes associated with both glycolysis and fatty acid oxidation. Conversely, neither increased extracellular urea nor UT-B inhibition had a significant impact on cell viability or cytotoxicity in SH-SY5Y neurones exposed to oxidative stressors tert-butyl hydroperoxide (t-BHP) and 6-hydroxydopamine (6-OHDA). Conclusions: This study further highlights the involvement of urea transport in regulating the neuroinflammation associated with AD. Moreover, we reveal a novel role for UT-B in maintaining microglial metabolic homeostasis. Taken together, these findings contribute supporting evidence to the regulation of UT-B as a therapeutic target for intervention into neuroinflammatory and neurodegenerative disease.
Collapse
Affiliation(s)
- Najlaa A. Al-Thani
- School of Biomolecular and Biomedical Science, University College Dublin, D04 V1W8 Dublin, Ireland; (N.A.A.-T.); (D.Z.)
- UCD Conway Institute, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Dylan Zinck
- School of Biomolecular and Biomedical Science, University College Dublin, D04 V1W8 Dublin, Ireland; (N.A.A.-T.); (D.Z.)
- UCD Conway Institute, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Gavin S. Stewart
- School of Biology and Environmental Science, University College Dublin, D04 V1W8 Dublin, Ireland;
| | - Derek A. Costello
- School of Biomolecular and Biomedical Science, University College Dublin, D04 V1W8 Dublin, Ireland; (N.A.A.-T.); (D.Z.)
- UCD Conway Institute, University College Dublin, D04 V1W8 Dublin, Ireland
| |
Collapse
|
43
|
Miao J, Lin J, Dong J, Amarasinghe O, Mason ER, Chu S, Qu Z, Cullers CC, Putt KS, Zhang ZY. Discovery and evaluation of novel SHIP-1 inhibitors. Bioorg Med Chem 2024; 114:117965. [PMID: 39454561 PMCID: PMC11551725 DOI: 10.1016/j.bmc.2024.117965] [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: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
Src Homology 2-containing Inositol 5'-Phosphatase-1 (SHIP-1), encoded by INPP5D, has been identified as an Alzheimer's disease (AD) risk-associated gene through recent genetic and epigenetic studies. SHIP-1 confers AD risk by inhibiting the TREM2 cascade and reducing beneficial microglial cellular processes, including phagocytosis. While several small molecules have been reported to modulate SHIP-1 activity, their limited selectivity and efficacy in advanced models restricted their potential as therapeutic agents or probes for biological studies. Herein, we validated and implemented a high-throughput screening platform to explore new chemotypes that can modulate the phosphatase activity of SHIP-1. We screened 49,260 central nervous system (CNS)-penetrate compounds sourced from commercial vendors using the malachite green-based assay for anti-SHIP-1 activity. Through analysis, prioritization, and validation of the screening hits, we identified three novel types of scaffolds that inhibit the SHIP-1 phosphatase activity with IC50s as low as 46.6 µM. To improve the inhibitory activity of these promising hits, we carried out structure-activity relationship (SAR) studies, resulting in a lead molecule SP3-12 that inhibits SHIP-1 with an IC50 value of 6.1 μM. Kinetic analyses of SP3-12 revealed that its inhibition mechanism is competitive, with a Ki value of 3.2 µM for SHIP-1 and a 7-fold selectivity over SHIP-2. Furthermore, results from testing in a microglial phagocytosis/cell health high content assay indicated that SP3-12 could effectively activate phagocytosis in human microglial clone 3 (HMC3) cells, with an EC50 of 2.0 µM, without cytotoxicity in the dose range. Given its potency, selectivity, and cellular activity, SP3-12 emerges as a promising small molecule inhibitor with potential for investigating the biological functions of SHIP-1.
Collapse
Affiliation(s)
- Jinmin Miao
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | - Jianping Lin
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | - Jiajun Dong
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | - Ovini Amarasinghe
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA
| | - Emily R Mason
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shaoyou Chu
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Zihan Qu
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Clayton C Cullers
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Karson S Putt
- Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA
| | - Zhong-Yin Zhang
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907, USA; Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA; Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
44
|
Yuan L, Song G, Xu W, Liu S, Zhang Y, Pan W, Ding X, Fu L, Lin Q, Sun F. Diethyl butylmalonate attenuates cognitive deficits and depression in 5×FAD mice. Front Neurosci 2024; 18:1480000. [PMID: 39588497 PMCID: PMC11586351 DOI: 10.3389/fnins.2024.1480000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/28/2024] [Indexed: 11/27/2024] Open
Abstract
Background Alzheimer's disease (AD), characterized by cognitive impairment and depression, is currently one of the intractable problems due to the insufficiency of intervention strategies. Diethyl butylmalonate (DBM) has recently attracted extensive interest due to its anti-inflammatory role in macrophages. However, it is still unknown whether DBM has a beneficial effect on cognitive deficits and depression. Methods DBM was administrated to 5×FAD and C57BL/6J mice by intraperitoneal injection. Novel object recognition, Y-maze spatial memory, Morris water maze and nest building tests were used to evaluate cognitive function. Moreover, the tail suspension test, forced swimming test, open field test and the elevated plus maze test were used to assess depression. Transmission electron microscopy, Golgi-Cox staining, immunofluorescence, RT-qPCR and western blot were utilized to determine the neuropathological changes in the hippocampus and amygdala of mice. Results Multiple behavioral tests showed that DBM effectively mitigated cognitive deficit and depression in 5×FAD mice. Moreover, DBM significantly attenuated synaptic ultrastructure and neurite impairment in the hippocampus of 5×FAD mice, paralleled by the improvement of the deficits of PSD95 and BDNF proteins. In addition, DBM decreased the accumulation of microglia and downregulated neuroinflammation in the hippocampus and amygdala of 5×FAD mice. Conclusion This study provides evidence that DBM ameliorates cognitive deficits and depression via improvement of the impairment of synaptic ultrastructure and neuroinflammation, suggesting that DBM is a potential drug candidate for treating AD-related neurodegeneration.
Collapse
Affiliation(s)
- Lai Yuan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, China
| | - Ge Song
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, China
| | - Wangwei Xu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
- Suqian Affiliated Hospital of Xuzhou Medical University, Suqian, China
| | - Shuni Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, China
| | - Yongsheng Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, China
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Xiaohui Ding
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Linlin Fu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Qisi Lin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Fenfen Sun
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
45
|
Kciuk M, Kruczkowska W, Gałęziewska J, Wanke K, Kałuzińska-Kołat Ż, Aleksandrowicz M, Kontek R. Alzheimer's Disease as Type 3 Diabetes: Understanding the Link and Implications. Int J Mol Sci 2024; 25:11955. [PMID: 39596023 PMCID: PMC11593477 DOI: 10.3390/ijms252211955] [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: 10/30/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Alzheimer's disease (AD) and type 2 diabetes mellitus (T2DM) are two prevalent conditions that present considerable public health issue in aging populations worldwide. Recent research has proposed a novel conceptualization of AD as "type 3 diabetes", highlighting the critical roles of insulin resistance and impaired glucose metabolism in the pathogenesis of the disease. This article examines the implications of this association, exploring potential new avenues for treatment and preventive strategies for AD. Key evidence linking diabetes to AD emphasizes critical metabolic processes that contribute to neurodegeneration, including inflammation, oxidative stress, and alterations in insulin signaling pathways. By framing AD within this metabolic context, we can enhance our understanding of its etiology, which in turn may influence early diagnosis, treatment plans, and preventive measures. Understanding AD as a manifestation of diabetes opens up the possibility of employing novel therapeutic strategies that incorporate lifestyle modifications and the use of antidiabetic medications to mitigate cognitive decline. This integrated approach has the potential to improve patient outcomes and deepen our comprehension of the intricate relationship between neurodegenerative diseases and metabolic disorders.
Collapse
Affiliation(s)
- Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland; (K.W.); (R.K.)
| | - Weronika Kruczkowska
- Department of Functional Genomics, Medical University of Lodz, 90-752 Lodz, Poland; (W.K.); (J.G.); (Ż.K.-K.)
| | - Julia Gałęziewska
- Department of Functional Genomics, Medical University of Lodz, 90-752 Lodz, Poland; (W.K.); (J.G.); (Ż.K.-K.)
| | - Katarzyna Wanke
- Department of Molecular Biotechnology and Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland; (K.W.); (R.K.)
| | - Żaneta Kałuzińska-Kołat
- Department of Functional Genomics, Medical University of Lodz, 90-752 Lodz, Poland; (W.K.); (J.G.); (Ż.K.-K.)
- Department of Biomedicine and Experimental Surgery, Medical University of Lodz, 90-136 Lodz, Poland
| | - Marta Aleksandrowicz
- Laboratory of Preclinical Research and Environmental Agents, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland;
| | - Renata Kontek
- Department of Molecular Biotechnology and Genetics, Faculty of Biology and Environmental Protection, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland; (K.W.); (R.K.)
| |
Collapse
|
46
|
Tenchov R, Sasso JM, Zhou QA. Alzheimer's Disease: Exploring the Landscape of Cognitive Decline. ACS Chem Neurosci 2024; 15:3800-3827. [PMID: 39392435 PMCID: PMC11587518 DOI: 10.1021/acschemneuro.4c00339] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/26/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, memory loss, and impaired daily functioning. The pathology of AD is marked by the accumulation of amyloid beta plaques and tau protein tangles in the brain, along with neuroinflammation and synaptic dysfunction. Genetic factors, such as mutations in APP, PSEN1, and PSEN2 genes, as well as the APOE ε4 allele, contribute to increased risk of acquiring AD. Currently available treatments provide symptomatic relief but do not halt disease progression. Research efforts are focused on developing disease-modifying therapies that target the underlying pathological mechanisms of AD. Advances in identification and validation of reliable biomarkers for AD hold great promise for enhancing early diagnosis, monitoring disease progression, and assessing treatment response in clinical practice in effort to alleviate the burden of this devastating disease. In this paper, we analyze data from the CAS Content Collection to summarize the research progress in Alzheimer's disease. We examine the publication landscape in effort to provide insights into current knowledge advances and developments. We also review the most discussed and emerging concepts and assess the strategies to combat the disease. We explore the genetic risk factors, pharmacological targets, and comorbid diseases. Finally, we inspect clinical applications of products against AD with their development pipelines and efforts for drug repurposing. The objective of this review is to provide a broad overview of the evolving landscape of current knowledge regarding AD, to outline challenges, and to evaluate growth opportunities to further efforts in combating the disease.
Collapse
Affiliation(s)
- Rumiana Tenchov
- CAS, a division of the American Chemical
Society, Columbus Ohio 43210, United States
| | - Janet M. Sasso
- CAS, a division of the American Chemical
Society, Columbus Ohio 43210, United States
| | | |
Collapse
|
47
|
Mayo P, Pascual J, Crisman E, Domínguez C, López MG, León R. Innovative pathological network-based multitarget approaches for Alzheimer's disease treatment. Med Res Rev 2024; 44:2367-2419. [PMID: 38678582 DOI: 10.1002/med.22045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/02/2024] [Accepted: 04/14/2024] [Indexed: 05/01/2024]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease and is a major health threat globally. Its prevalence is forecasted to exponentially increase during the next 30 years due to the global aging population. Currently, approved drugs are merely symptomatic, being ineffective in delaying or blocking the relentless disease advance. Intensive AD research describes this disease as a highly complex multifactorial disease. Disclosure of novel pathological pathways and their interconnections has had a major impact on medicinal chemistry drug development for AD over the last two decades. The complex network of pathological events involved in the onset of the disease has prompted the development of multitarget drugs. These chemical entities combine pharmacological activities toward two or more drug targets of interest. These multitarget-directed ligands are proposed to modify different nodes in the pathological network aiming to delay or even stop disease progression. Here, we review the multitarget drug development strategy for AD during the last decade.
Collapse
Affiliation(s)
- Paloma Mayo
- Departamento de desarrollo preclínico, Fundación Teófilo Hernando, Las Rozas, Madrid, Spain
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Jorge Pascual
- Departamento de desarrollo preclínico, Fundación Teófilo Hernando, Las Rozas, Madrid, Spain
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Enrique Crisman
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Cristina Domínguez
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Manuela G López
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Rafael León
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| |
Collapse
|
48
|
Xie H, Wu F, Mao J, Wang Y, Zhu J, Zhou X, Hong K, Li B, Qiu X, Wen C. The role of microglia in neurological diseases with involvement of extracellular vesicles. Neurobiol Dis 2024; 202:106700. [PMID: 39401551 DOI: 10.1016/j.nbd.2024.106700] [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: 08/28/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/20/2024] Open
Abstract
As a subset of mononuclear phagocytes in the central nervous system, microglia play a crucial role in immune defense and homeostasis maintenance. Microglia can regulate their states in response to specific signals of health and pathology. Microglia-mediated neuroinflammation is a pathological hallmark of neurodegenerative diseases, neurological damage and neurological tumors, underscoring its key immunoregulatory role in these conditions. Intriguingly, a substantial body of research has indicated that extracellular vesicles can mediate intercellular communication by transporting cargoes from parental cells, a property that is also reflected in microenvironmental signaling networks involving microglia. Based on the microglial characteristics, we briefly outline the biological features of extracellular vesicles and focus on summarizing the integrative role played by microglia in the maintenance of nervous system homeostasis and progression of different neurological diseases. Extracellular vesicles may engage in the homeostasis maintenance and pathological process as a medium of intercellular communication. Here, we aim to provide new insights for further exploration of neurological disease pathogenesis, which may provide theoretical foundations for cell-free therapies.
Collapse
Affiliation(s)
- Haotian Xie
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Feifeng Wu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Jueyi Mao
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yang Wang
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Junquan Zhu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Xin Zhou
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Kimsor Hong
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Binbin Li
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Xinying Qiu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Chuan Wen
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha 410011, China.
| |
Collapse
|
49
|
Dadu A, Ta M, Tustison NJ, Daneshmand A, Marek K, Singleton AB, Campbell RH, Nalls MA, Iwaki H, Avants B, Faghri F. Prediction, prognosis and monitoring of neurodegeneration at biobank-scale via machine learning and imaging. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.27.24316215. [PMID: 39574848 PMCID: PMC11581077 DOI: 10.1101/2024.10.27.24316215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Background Alzheimer's disease and related dementias (ADRD) and Parkinson's disease (PD) are the most common neurodegenerative conditions. These central nervous system disorders impact both the structure and function of the brain and may lead to imaging changes that precede symptoms. Patients with ADRD or PD have long asymptomatic phases that exhibit significant heterogeneity. Hence, quantitative measures that can provide early disease indicators are necessary to improve patient stratification, clinical care, and clinical trial design. This work uses machine learning techniques to derive such a quantitative marker from T1-weighted (T1w) brain Magnetic resonance imaging (MRI). Methods In this retrospective study, we developed machine learning (ML) based disease-specific scores based on T1w brain MRI utilizing Parkinson's Disease Progression Marker Initiative (PPMI) and Alzheimer's Disease Neuroimaging Initiative (ADNI) cohorts. We evaluated the potential of ML-based scores for early diagnosis, prognosis, and monitoring of ADRD and PD in an independent large-scale population-based longitudinal cohort, UK Biobank. Findings 1,826 dementia images from 731 participants, 3,161 healthy control images from 925 participants from the ADNI cohort, 684 PD images from 319 participants, and 232 healthy control images from 145 participants from the PPMI cohort were used to train machine learning models. The classification performance is 0.94 [95% CI: 0.93-0.96] area under the ROC Curve (AUC) for ADRD detection and 0.63 [95% CI: 0.57-0.71] for PD detection using 790 extracted structural brain features. The most predictive regions include the hippocampus and temporal brain regions in ADRD and the substantia nigra in PD. The normalized ML model's probabilistic output (ADRD and PD imaging scores) was evaluated on 42,835 participants with imaging data from the UK Biobank. There are 66 cases for ADRD and 40 PD cases whose T1 brain MRI is available during pre-diagnostic phases. For diagnosis occurrence events within 5 years, the integrated survival model achieves a time-dependent AUC of 0.86 [95% CI: 0.80-0.92] for dementia and 0.89 [95% CI: 0.85-0.94] for PD. ADRD imaging score is strongly associated with dementia-free survival (hazard ratio (HR) 1.76 [95% CI: 1.50-2.05] per S.D. of imaging score), and PD imaging score shows association with PD-free survival (hazard ratio 2.33 [95% CI: 1.55-3.50]) in our integrated model. HR and prevalence increased stepwise over imaging score quartiles for PD, demonstrating heterogeneity. As a proxy for diagnosis, we validated AD/PD polygenic risk scores of 42,835 subjects against the imaging scores, showing a highly significant association after adjusting for covariates. In both the PPMI and ADNI cohorts, the scores are associated with clinical assessments, including the Mini-Mental State Examination (MMSE), Alzheimer's Disease Assessment Scale-cognitive subscale (ADAS-Cog), and pathological markers, which include amyloid and tau. Finally, imaging scores are associated with polygenic risk scores for multiple diseases. Our results suggest that we can use imaging scores to assess the genetic architecture of such disorders in the future. Interpretation Our study demonstrates the use of quantitative markers generated using machine learning techniques for ADRD and PD. We show that disease probability scores obtained from brain structural features are useful for early detection, prognosis prediction, and monitoring disease progression. To facilitate community engagement and external tests of model utility, an interactive app to explore summary level data from this study and dive into external data can be found here https://ndds-brainimaging-ml.streamlit.app. As far as we know, this is the first publicly available cloud-based MRI prediction application. Funding US National Institute on Aging, and US National Institutes of Health.
Collapse
Affiliation(s)
- Anant Dadu
- Department of Computer Science, University of Illinois at Urbana-Champaign, Champaign, IL, 61820, USA
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, 20892, USA
- DataTecnica, Washington, DC, 20812, USA
| | - Michael Ta
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, 20892, USA
- DataTecnica, Washington, DC, 20812, USA
| | - Nicholas J Tustison
- University of Virginia, Dept of Radiology and Medical Imaging, Charlottesville, VA, 22903, USA
| | - Ali Daneshmand
- Department of Neurology, Boston Medical Center, Boston University School of Medicine, Boston, MA, 02118, USA
| | | | - Andrew B Singleton
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, 20892, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Roy H Campbell
- Department of Computer Science, University of Illinois at Urbana-Champaign, Champaign, IL, 61820, USA
| | - Mike A Nalls
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, 20892, USA
- DataTecnica, Washington, DC, 20812, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Hirotaka Iwaki
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, 20892, USA
- DataTecnica, Washington, DC, 20812, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Brian Avants
- University of Virginia, Dept of Radiology and Medical Imaging, Charlottesville, VA, 22903, USA
- InviCRO LLC, Boston, Massachusetts
| | - Faraz Faghri
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, 20892, USA
- DataTecnica, Washington, DC, 20812, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
50
|
Zhang L, Lin J, Xiang K, Shi T, Guo B. Omnidirectional improvement of mitochondrial health in Alzheimer's disease by multi-targeting engineered activated neutrophil exosomes. J Control Release 2024; 376:470-487. [PMID: 39433157 DOI: 10.1016/j.jconrel.2024.10.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/14/2024] [Accepted: 10/17/2024] [Indexed: 10/23/2024]
Abstract
Alzheimer's disease (AD) is one kind of devasting neurodegenerative disorders affecting over 50 million people worldwide. Multi-targeted therapy has emerged as a new treatment for diagnosing and alleviating the pathogenesis process of AD; however, the current strategy is limited by its unsatisfactory efficiency. In our study, engineered activated neutrophil-derived exosomes (MP@Cur-MExo) were developed to improve the mitochondrial function in neurons by targeting and alleviating Aβ-induced neurotoxicity. MP@Cur-MExo are exosomes derived from IL-8-stimulated neutrophils decorated with mitochondria targeting ligand and Aβ targeted ligand modified SPION. Engineered exosomes can be cleaved by matrix metallopeptidase-2, which is overexpressed in the AD brain. Consequently, the released SPION and Curcumin-loaded engineered exosomes collaboratively protected neuron cells against Aβ-induced mitochondrial deficiency. In addition, MP@Cur-MExo effectively accumulated in the inflamed region of AD brain at an early stage, allowing early diagnosis of AD through bimodal (MRI/IVIS) imaging. Importantly, in a mouse model at an early stage of AD, intravenously injected MP@Cur-MExo restored mitochondrial function and reduced Aβ-induced mitochondrial damage, thereby attenuating AD progression. In conclusion, our designed engineered exosomes demonstrated that omnidirectional improvement of mitochondrial function can serve as a novel and practical approach for the diagnosis and treatment of neurodegenerative diseases. This study also reveals a promising therapeutic agent for impeding AD progression for future clinical applications.
Collapse
Affiliation(s)
- Lei Zhang
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China.
| | - Jiaquan Lin
- Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Branch of National Clinical Research Center for Orthopedics Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu 210008, China
| | - Kai Xiang
- School of Pharmacy, Wannan Medical College, Wuhu 241002, China
| | - Tianshu Shi
- Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Branch of National Clinical Research Center for Orthopedics Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu 210008, China.
| | - Baosheng Guo
- Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Branch of National Clinical Research Center for Orthopedics Sports Medicine and Rehabilitation, 321 Zhongshan Road, Nanjing, Jiangsu 210008, China.
| |
Collapse
|