1
|
Paitel ER, Pettigrew C, Moghekar A, Miller MI, Faria AV, Albert M, Soldan A. Alzheimer's disease cerebrospinal fluid biomarker levels and APOE genetic status are associated with hippocampal-cerebellar functional connectivity. Neurobiol Aging 2025; 151:107-116. [PMID: 40273528 PMCID: PMC12101073 DOI: 10.1016/j.neurobiolaging.2025.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/20/2025] [Accepted: 04/12/2025] [Indexed: 04/26/2025]
Abstract
Recent research suggests that hippocampal-cerebellar (Hp-CB) functional connectivity may be altered early in the course of Alzheimer's disease (AD), given the early accumulation of AD pathology in the hippocampi and emerging evidence of cerebellar changes in early AD. This study analyzed the role of AD genetic risk (via APOE ε4 carrier status) and cerebrospinal fluid (CSF) biomarkers of AD pathology (ratio of phosphorylated tau (p-tau181) to amyloid beta (Aβ42/Aβ40)) on the relationship between age and functional Hp-CB resting state fMRI connectivity in 161 cognitively unimpaired older adults (M age =67.3; SD =9.0; 37 % APOE ε4 +). In multiple regression analyses with Hp-CB connectivity as the outcome, there were significant interactions between age and APOE ε4 status, and between age and CSF AD biomarkers. Older age was associated with greater Hp-CB connectivity in APOE ε4 non-carriers and participants with less abnormal CSF AD biomarkers. In contrast, Hp-CB connectivity was marginally lower with older age in ε4 carriers and those with more abnormal AD biomarkers. Furthermore, greater Hp-CB connectivity was associated with better episodic memory performance across all groups. These findings suggest that age-related increases in Hp-CB connectivity among APOE ε4 non-carriers and those with low AD biomarker levels reflect age-related changes that are largely unrelated to AD, while age-related decreases in Hp-CB connectivity in APOE ε4 carriers may reflect AD-related alterations. These findings also highlight the importance of cerebellar contributions to cognitive performance among older adults and suggest that Hp-CB connectivity may be altered in preclinical AD.
Collapse
Affiliation(s)
- Elizabeth R Paitel
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Corinne Pettigrew
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael I Miller
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Andreia V Faria
- Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
| | - Marilyn Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anja Soldan
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
2
|
Khan S, Iqbal T, Rehman MU, Khan MB, Islam MS, Dahlous KA. Integrated insight and in silico investigation of hybrid bis-thiazolidinone derivatives along with anti-Alzheimer activity. 3 Biotech 2025; 15:176. [PMID: 40390986 PMCID: PMC12085477 DOI: 10.1007/s13205-025-04313-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 04/05/2025] [Indexed: 06/03/2025] Open
Abstract
UNLABELLED In the current study, a novel route was established for the synthesis of hybrid benzothiazole derived thiazole bearing bis-thiazolidinone-chalcone (1-15) scaffolds. These compounds were screened for their biological potential as anti-Alzheimer therapeutic agents by inhibiting acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) enzymes. The biological evaluation and molecular docking studies revealed that most of the synthesized compounds exhibited significant inhibitory activity against both enzymes, outperforming the standard drug, donepezil. Among them, Analog 15 demonstrated remarkable therapeutic potential, with IC₅₀ values of 3.30 ± 0.70 µM and 3.80 ± 0.90 µM, as well as strong binding affinities/docking scores of - 8.97 and - 12.84 kcal/mol for AChE and BuChE, respectively. Additionally, enzyme kinetics analysis using Lineweaver-Burk plots confirmed the mode of inhibition of the synthesized analogs. Pharmacokinetic predictions further supported the drug-like properties of these compounds, highlighting favorable pharmacological profiles, including good water solubility, non-carcinogenicity, and biological safety. The findings presented in this study provide compelling evidence for the anti-Alzheimer potential of these novel scaffolds, warranting further investigation through in vivo studies and clinical exploration to assess their full therapeutic applicability. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s13205-025-04313-6.
Collapse
Affiliation(s)
- Shoaib Khan
- Department of Chemistry, Abbottabad University of Science and Technology, Abbottabad, 22500 Pakistan
| | - Tayyiaba Iqbal
- Department of Chemistry, Abbottabad University of Science and Technology, Abbottabad, 22500 Pakistan
| | - Mujaddad Ur Rehman
- Department of Microbiology, Abbottabad University of Science and Technology, Abbottabad, 22500 Pakistan
| | - Muhammad Bilal Khan
- Department of Chemistry, Abbottabad University of Science and Technology, Abbottabad, 22500 Pakistan
| | - Mohammad Shahidul Islam
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, 11451 Riyadh, Saudi Arabia
| | - Kholood A. Dahlous
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, 11451 Riyadh, Saudi Arabia
| |
Collapse
|
3
|
González-Escalante A, Milà-Alomà M, Brum WS, Ashton NJ, Ortiz-Romero P, Shekari M, Campo MD, Anastasi F, Quijano-Rubio C, Kollmorgen G, Minguillón C, Sánchez-Benavides G, Grau-Rivera O, Gispert JD, Zetterberg H, Vilor-Tejedor N, Blennow K, Suárez-Calvet M. A plasma biomarker panel for detecting early amyloid-β accumulation and its changes in middle-aged cognitively unimpaired individuals at risk for Alzheimer's disease. EBioMedicine 2025; 116:105741. [PMID: 40414160 PMCID: PMC12159936 DOI: 10.1016/j.ebiom.2025.105741] [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: 10/11/2024] [Revised: 04/16/2025] [Accepted: 04/22/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND Plasma biomarkers of Alzheimer's disease (AD) change during preclinical stages, indicating potential for detecting amyloid-β (Aβ) pathology in cognitively unimpaired (CU) individuals. Given the need for accurate, scalable biomarkers, we evaluated a fully automated plasma panel to detect and monitor longitudinal Aβ accumulation in CU individuals. METHODS In this longitudinal study, we examined a plasma panel (Aβ42/40, p-tau181, GFAP, NfL, p-tau217 and ApoE4) in CU participants at risk for AD. We assessed the biomarkers' performance to detect Aβ pathology and the cross-sectional and longitudinal relationships between the biomarkers and Aβ accumulation, neurodegeneration and cognition. FINDINGS We included 400 middle-aged CU participants, of whom 135 (33.8%) were CSF Aβ-positive. All plasma biomarkers differed between Aβ-positive and -negative individuals, with plasma Aβ42/40, p-tau217, p-tau181/Aβ42, and p-tau217/Aβ42 showing the best performance in detecting A+ CU individuals. However, plasma Aβ42/40 was sensitive to random variability. Plasma p-tau217/Aβ42 had the highest performance in detecting PET A+ individuals (AUC = 0.94). All baseline plasma biomarkers were associated with longitudinal increases in Aβ deposition (mean follow-up [SD]: 3.27 ± 0.5). Longitudinal changes in plasma p-tau217 and p-tau217/Aβ42 were associated with concurrent changes in Aβ (both CSF and PET) and soluble tau pathology. INTERPRETATION In CU individuals, several plasma biomarkers at baseline detect Aβ accumulation and are associated with its short-term change. Plasma p-tau217, and p-tau217/Aβ42 longitudinal changes reflect concurrent Aβ accumulation during this period. These findings help enrich studies in CU individuals at risk of progressing to AD. FUNDING ERC-2020-STG (Grant agreement No. 948677); ERA PerMed-ERA NET and the Generalitat de Catalunya (SLD077/21/000001); PI19/00155; PI22/00456, LCF/BQ/PR21/11840004.
Collapse
Affiliation(s)
- Armand González-Escalante
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Hospital del Mar Research Institute, Barcelona, Spain; Universitat Pompeu Fabra, Barcelona, Spain
| | - Marta Milà-Alomà
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, United States of America; Department of Veterans Affairs Medical Center, Northern California Institute for Research and Education (NCIRE), San Francisco, CA, United States of America
| | - Wagner S Brum
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, United Kingdom; Banner Alzheimer's Institute and University of Arizona, Phoenix, AZ, United States of America; Banner Sun Health Research Institute, Sun City, AZ, United States of America
| | - Paula Ortiz-Romero
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Hospital del Mar Research Institute, Barcelona, Spain
| | - Mahnaz Shekari
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Hospital del Mar Research Institute, Barcelona, Spain
| | - Marta Del Campo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Hospital del Mar Research Institute, Barcelona, Spain
| | - Federica Anastasi
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Hospital del Mar Research Institute, Barcelona, Spain; Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | | | | | - Carolina Minguillón
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Hospital del Mar Research Institute, Barcelona, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Gonzalo Sánchez-Benavides
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Hospital del Mar Research Institute, Barcelona, Spain
| | - Oriol Grau-Rivera
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Hospital del Mar Research Institute, Barcelona, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain; Servei de Neurologia, Hospital del Mar, Barcelona, Spain
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Hospital del Mar Research Institute, Barcelona, Spain; Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Madrid, Spain; Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, Dementia Research Centre, UCL Institute of Neurology, London, United Kingdom; UK Dementia Research Institute, University College London, London, United Kingdom; Hong Kong Center for Neurodegenerative Diseases (HKCeND), Hong Kong, China; Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Natalia Vilor-Tejedor
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Hospital del Mar Research Institute, Barcelona, Spain; Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; Department of Genetics, Radboud University Nijmegen, Nijmegen, Netherlands
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France; Division of Life Sciences and Medicine and Department of Neurology, Neurodegenerative Disorder Research Center, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, PR China
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain; Hospital del Mar Research Institute, Barcelona, Spain; Servei de Neurologia, Hospital del Mar, Barcelona, Spain.
| |
Collapse
|
4
|
Raj A, Torok J, Ranasinghe K. Understanding the complex interplay between tau, amyloid and the network in the spatiotemporal progression of Alzheimer's disease. Prog Neurobiol 2025; 249:102750. [PMID: 40107380 DOI: 10.1016/j.pneurobio.2025.102750] [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: 10/04/2024] [Revised: 02/24/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
INTRODUCTION The interaction of amyloid and tau in neurodegenerative diseases is a central feature of AD pathophysiology. While experimental studies point to various interaction mechanisms, their causal direction and mode (local, remote or network-mediated) remain unknown in human subjects. The aim of this study was to compare mathematical reaction-diffusion models encoding distinct cross-species couplings to identify which interactions were key to model success. METHODS We tested competing mathematical models of network spread, aggregation, and amyloid-tau interactions on publicly available data from ADNI. RESULTS Although network spread models captured the spatiotemporal evolution of tau and amyloid in human subjects, the model including a one-way amyloid-to-tau aggregation interaction performed best. DISCUSSION This mathematical exposition of the "pas de deux" of co-evolving proteins provides quantitative, whole-brain support to the concept of amyloid-facilitated-tauopathy rather than the classic amyloid-cascade or pure-tau hypotheses, and helps explain certain known but poorly understood aspects of AD.
Collapse
Affiliation(s)
- Ashish Raj
- Department of Radiology, University of California at San Francisco, USA; Bakar Computational Health Sciences Institute, UCSF, USA.
| | - Justin Torok
- Department of Radiology, University of California at San Francisco, USA
| | - Kamalini Ranasinghe
- The Memory and Aging Center, Department of Neurology, University of California at San Francisco, USA
| |
Collapse
|
5
|
Sabbagh MN, Zhao C, Mahendran M, Jang SR, Laliberté F, Toyosaki H, Zhang K, Frech F, Nair KV. Characterizing the Journey of Early Alzheimer's Disease in Patients Initiating Lecanemab Treatment in the United States: A Real-World Evidence Study. Neurol Ther 2025; 14:1115-1127. [PMID: 40319433 PMCID: PMC12089008 DOI: 10.1007/s40120-025-00756-4] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Accepted: 04/15/2025] [Indexed: 05/07/2025] Open
Abstract
INTRODUCTION With the advent of disease-modifying therapies for early Alzheimer's disease (AD), a comprehensive characterization of patients initiating lecanemab in the USA is needed to understand its use in real-world settings. METHODS This retrospective observational study used administrative claims from the Komodo Research Database (1/1/2023-6/30/2024). Eligible patients had ≥ 1 lecanemab administration (first claim defined the index date) and ≥ 12 months of clinical activity/insurance eligibility before the index date. Patient characteristics, diagnostic process, and AD-related medications were evaluated within 12 months before the index date (baseline), whereas lecanemab treatment patterns and concomitant medications were evaluated on or after the index date (follow-up). Outcomes were reported using descriptive statistics and persistence to lecanemab was evaluated using Kaplan-Meier analysis. RESULTS Of 3155 patients included in the study, mean age was 75.0 years, 55.8% were female, 44.2% were male, and most (93.3%) received their index lecanemab administration in an urban setting. Diagnoses of AD (83.8%) and mild cognitive impairment (60.8%) were common at baseline, and 67.6% of patients used AD symptomatic medications. Average time from earliest diagnosis to first lecanemab administration was 4.9 months among patients with a diagnosis in January 2023 (accelerated approval date) or onwards. Over a mean follow-up of 138.8 days, the monthly mean number of administrations of lecanemab was 1.9, with an average of 16.5 days between consecutive administrations and 47.4 days to the first follow-up head magnetic resonance imaging. Persistence to lecanemab was 87.6% at 4 months after treatment initiation. CONCLUSION Lecanemab was utilized in appropriate patient populations according to the prescribing information approved by the US Food and Drug Administration. Findings from our study provide first insights into the real-world use of lecanemab in the USA and shed light on the need for increased and timely lecanemab initiation for the long-term management of early AD.
Collapse
Affiliation(s)
- Marwan N Sabbagh
- Department of Neurology, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Chenyue Zhao
- Eisai Inc., 200 Metro Blvd, Nutley, NJ, 07110, USA.
| | | | | | | | | | | | - Feride Frech
- Eisai Inc., 200 Metro Blvd, Nutley, NJ, 07110, USA
| | - Kavita V Nair
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
6
|
Yan F, Qiao Y, Pan S, Kang A, Chen H, Bai Y. RIPK1: A Promising Target for Intervention Neuroinflammation. J Neuroimmune Pharmacol 2025; 20:59. [PMID: 40418439 DOI: 10.1007/s11481-025-10208-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/16/2025] [Indexed: 05/27/2025]
Abstract
Necroptosis is a novel mode of cell death that differs from traditional apoptosis, characterized by distinct molecular mechanisms and physiopathological features. Recent research has increasingly underscored the pivotal role of necroptosis in various neurological diseases, including stroke, Alzheimer's disease and multiple sclerosis. A defining hallmark of these conditions is neuroinflammation, a complex inflammatory response that critically influences neuronal survival. This review provides a comprehensive analysis of the mechanistic underpinnings of necroptosis and its intricate interplay with neuroinflammation, exploring the interrelationship between the two processes and their impact on neurological disorders. In addition, we discuss potential therapeutic strategies that target the intervention of necroptosis and neuroinflammation, offering novel avenues for intervention. By deepening our understanding of these interconnected processes, the development of more effective treatments approaches holds significant promise for improving patient outcomes in neurological disorders.
Collapse
Affiliation(s)
- Feixing Yan
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Yujun Qiao
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Shunli Pan
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Anjuan Kang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Haile Chen
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Yinliang Bai
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, China.
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, 730000, China.
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, 730000, China.
| |
Collapse
|
7
|
Li Y, Yao YL, Wu Y. Causal relationships between plasma proteins and Alzheimer's disease using bidirectional Mendelian randomization. J Alzheimers Dis 2025:13872877251345151. [PMID: 40397384 DOI: 10.1177/13872877251345151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
BackgroundAlzheimer's disease (AD) is influenced by a complex interplay of genetic, immune, and metabolic factors. Identifying plasma proteins causally linked to AD could help clarify these pathways and uncover potential therapeutic targets.ObjectiveThis study aims to investigate the causal relationships between AD and plasma proteins.MethodsWe conducted a two-stage, two-sample Mendelian randomization (MR) analysis to explore the causal relationships between plasma protein levels and AD risk. In both stages, we used non-overlapping genome-wide association study datasets for exposures (plasma protein levels) and outcome (AD) to ensure robust and independent analyses. We examined both forward (from plasma proteins to AD risk) and reverse (from AD to plasma protein expression) causal effects to elucidate potential bidirectional relationships.ResultsOur MR analysis identified 25 plasma proteins with causal associations to AD, with many implicated in immune and lipid metabolic pathways. These findings reinforce the roles of inflammation and lipid metabolism in AD pathogenesis and offer novel insights into specific proteins that may serve as biomarkers or therapeutic targets.ConclusionsThis study provides further support for the relationship between immune and lipid metabolic dysregulation and AD, advancing our understanding of the molecular mechanisms underlying disease progression and highlighting key proteins for future research and therapeutic development.
Collapse
Affiliation(s)
- Yichen Li
- Department of Psychiatry, Wuhan Mental Health Center, Wuhan, Hubei, China
- Affiliated Wuhan Mental Health Center, Jianghan University, Wuhan, Hubei, China
| | - Yu-Lin Yao
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, Hubei, China
| | - Yong Wu
- Department of Psychiatry, Wuhan Mental Health Center, Wuhan, Hubei, China
- Affiliated Wuhan Mental Health Center, Jianghan University, Wuhan, Hubei, China
| |
Collapse
|
8
|
Fu Y, Zhang J, Yang C, Wang Y, Yang Y, Qiu P, Xie W, Zhang S, Lǚ T. Effects of Solvent Dimethyl Sulfoxide Invites a Rethink of Its Application in Amyloid Beta Cytotoxicity. Int J Toxicol 2025:10915818251338235. [PMID: 40373217 DOI: 10.1177/10915818251338235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2025]
Abstract
Dimethyl sulfoxide (DMSO) is commonly used as a solvent for preparing amyloid-beta (Aβ) peptides implicated in Alzheimer's disease. While considered relatively non-toxic at low concentrations, DMSO itself may exert biological effects that could confound experimental outcomes, especially for weakly cytotoxic substances like Aβ. Seven brain cell types (BV-2, N2a, SH-SY5Y, U87, neurons, astrocytes, microglia) were treated with varying DMSO concentrations or Aβ1-42 oligomers/protofibrils/fibrils prepared using DMSO. Cell viability was assessed by CCK-8 and LDH assays. Matched DMSO controls were prepared alongside Aβ treatments to delineate solvent effects. Low DMSO concentrations (0.0625-0.015625%) exhibited hormetic cytoprotective and growth-promoting effects, while higher concentrations (≥2%) were cytotoxic. Importantly, these hormetic solvent effects confounded the measurement of Aβ cytotoxicity. By accounting for matched DMSO controls, the study revealed that Aβ fibril toxicity may have been underestimated due to the cytoprotective solvent effects of low DMSO concentrations used in their preparation. In conclusion, DMSO exhibits complex hormetic dose-responses that can significantly influence experimental outcomes, especially for weakly cytotoxic agents like Aβ. Rigorous solvent controls are crucial to delineate genuine substance effects from potential solvent confounds and avoid erroneous interpretations.
Collapse
Affiliation(s)
- Yanhong Fu
- Department of Neurology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Center for Cognition and Sleep, The People's Hospital of Guangxi Zhuang Autonomous Region & Institute of Brain and Mental Diseases, Guangxi Academy of Medical Sciences, Nanning, China
| | - Jiafa Zhang
- Department of Neurology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Canhong Yang
- Department of Neurology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Yuanyuan Wang
- Department of Neurology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Yunzhu Yang
- Department of Neurology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Pingming Qiu
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Weibing Xie
- School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Shufen Zhang
- Internal Medicine Department, The Second People's Hospital of Guangzhou Nansha, Guangzhou, China
| | - Tianming Lǚ
- Department of Neurology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
9
|
Begh MZA, Zehravi M, Bhuiyan MAK, Molla MR, Raman K, Emran TB, Ullah MH, Ahmad I, Osman H, Khandaker MU. Recent advances in stem cell approaches to neurodegeneration: A comprehensive review with mechanistic insight. Pathol Res Pract 2025; 271:156013. [PMID: 40381433 DOI: 10.1016/j.prp.2025.156013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2025] [Revised: 05/10/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025]
Abstract
The progressive nature of neurodegenerative diseases (NDs), such as Parkinson's disease, Alzheimer's disease, Huntington's disease, and amyotrophic lateral sclerosis, presents substantial problems because current treatments are still obscure. Stem cell-based treatments are emerging as a viable solution to address the significant gaps in treating these severe diseases. This study provides a comprehensive analysis of the latest advancements in stem cell research, focusing on the treatment of NDs. Various types of stem cells, such as adult, induced pluripotent, and embryonic stem cells, and their potential applications in immunomodulation, neurotrophic factor release, and neuronal development are also discussed. Recent clinical studies reveal outcomes, challenges, and solutions, with advancements in disease-specific neural cell production, gene editing, and improved stem cell transplantation transport strategies. The review discussed future perspectives on developing more effective stem cell-based interventions. Biomaterials are being used for cell distribution and personalized medicine techniques to improve treatment outcomes, while exploring stem cell treatments for NDs and identifying areas for further research.
Collapse
Affiliation(s)
- Md Zamshed Alam Begh
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka 1216, Bangladesh.
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy, College of Dentistry & Pharmacy, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia.
| | | | - M Raju Molla
- Department of Pharmacy, Atish Dipankar University of Science and Technology, Dhaka 1230, Bangladesh
| | - Kannan Raman
- Department of Pharmacology, St. John's College of Pharmaceutical Sciences & Research, Kattappana, Idukki, Kerala, India
| | - Talha Bin Emran
- Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka 1216, Bangladesh
| | - Md Habib Ullah
- Department of Physics, American International University-Bangladesh (AIUB), 408/1, Kuratoli, Khilkhet, Dhaka 1229, Bangladesh
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Hamid Osman
- Department of Radiological Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia
| | - Mayeen Uddin Khandaker
- Applied Physics and Radiation Technologies Group, CCDCU, Faculty of Engineering and Technology, Sunway University, Bandar Sunway, 47500 Selangor, Malaysia; Department of Physics, College of Science, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| |
Collapse
|
10
|
Papanikolaou A, Graykowski D, Lee BI, Yang M, Ellingford R, Zünkler J, Bond SA, Rowland JM, Rajani RM, Harris SS, Sharp DJ, Busche MA. Selectively vulnerable deep cortical layer 5/6 fast-spiking interneurons in Alzheimer's disease models in vivo. Neuron 2025:S0896-6273(25)00293-4. [PMID: 40345184 DOI: 10.1016/j.neuron.2025.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 03/03/2025] [Accepted: 04/11/2025] [Indexed: 05/11/2025]
Abstract
Alzheimer's disease (AD) is initiated by amyloid-beta (Aβ) accumulation in the neocortex; however, the cortical layers and neuronal cell types first susceptible to Aβ remain unknown. Using in vivo two-photon Ca2+ imaging in the visual cortex of AD mouse models, we found that cortical layer 5 neurons displayed abnormally prolonged Ca2+ transients before substantial plaque formation. Neuropixels recordings revealed that these abnormal transients were associated with reduced spiking and impaired visual tuning of parvalbumin (PV)-positive fast-spiking interneurons (FSIs) in layers 5/6, whereas PV-FSIs in superficial layers remained unaffected. These dysfunctions occurred alongside a deep-layer-specific reduction in neuronal pentraxin 2 (NPTX2) within excitatory neurons, decreased GluA4 in PV-FSIs, and fewer excitatory synapses onto PV-FSIs. Notably, NPTX2 overexpression increased excitatory input onto layers 5/6 PV-FSIs and rectified their spiking activity. Thus, our findings reveal an early selective impairment of deep cortical layers 5/6 in AD models and identify deep-layer PV-FSIs as therapeutic targets.
Collapse
Affiliation(s)
| | - David Graykowski
- UK Dementia Research Institute at University College London, London, UK
| | - Byung Il Lee
- UK Dementia Research Institute at University College London, London, UK
| | - Mengke Yang
- UK Dementia Research Institute at University College London, London, UK
| | - Robert Ellingford
- UK Dementia Research Institute at University College London, London, UK
| | - Jana Zünkler
- UK Dementia Research Institute at University College London, London, UK
| | - Suraya A Bond
- UK Dementia Research Institute at University College London, London, UK
| | - James M Rowland
- UK Dementia Research Institute at University College London, London, UK
| | - Rikesh M Rajani
- UK Dementia Research Institute at University College London, London, UK; British Heart Foundation - UK Dementia Research Institute Centre for Vascular Dementia Research at The University of Edinburgh, Edinburgh, UK
| | - Samuel S Harris
- UK Dementia Research Institute at University College London, London, UK
| | - David J Sharp
- UK Dementia Research Institute Care Research & Technology Centre and Department of Brain Sciences, Imperial College London, London, UK
| | - Marc Aurel Busche
- UK Dementia Research Institute at University College London, London, UK; Department of Neurodegenerative Diseases, University Hospital of Geriatric Medicine FELIX PLATTER and University of Basel, Basel, Switzerland; Department of Biomedicine, University of Basel, Basel, Switzerland.
| |
Collapse
|
11
|
Weiner S, Sauer M, Montoliu-Gaya L, Benedet AL, Ashton NJ, Gonzalez-Ortiz F, Simrén J, Rahmouni N, Tissot C, Therriault J, Servaes S, Stevenson J, Leinonen V, Rauramaa T, Hiltunen M, Rosa-Neto P, Blennow K, Zetterberg H, Gobom J. Cerebrospinal fluid proteome profiling across the Alzheimer's disease continuum: a step towards solving the equation for 'X'. Mol Neurodegener 2025; 20:52. [PMID: 40329321 PMCID: PMC12057231 DOI: 10.1186/s13024-025-00841-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND While the temporal profile of amyloid (Aβ) and tau cerebrospinal fluid (CSF) biomarkers along the Alzheimer's disease (AD) continuum is well-studied, chronological changes of CSF proteins reflecting other disease-relevant processes, denoted 'X' in the ATX(N) framework, remain poorly understood. METHODS Using an untargeted mass spectrometric approach termed tandem mass tag (TMT), we quantified over 1500 CSF proteins across the AD continuum in three independent cohorts, finely staged by Aβ/tau positron emission tomography (PET), fluid biomarkers, or brain biopsy. Weighted protein co-expression network analysis identified clusters of proteins robustly correlating in all three cohorts which sequentially changed with AD progression. Obtained protein clusters were correlated with fluid biomarker measurements (phosphorylated tau (p-tau) species including p-tau181, p-tau217, and p-tau205, as well as Aβ), Aβ/tau PET imaging, and clinical parameters to discern disease-relevant clusters which were modelled across the AD continuum. RESULTS Neurodegeneration-related proteins (e.g., 14-3-3 proteins, PPIA), derived from different brain cell types, strongly correlated with fluid as well as imaging biomarkers and increased early in the AD continuum. Among them, the proteins SMOC1 and CNN3 were highly associated with Aβ pathology, while the 14-3-3 proteins YWHAZ and YWHAE as well as PPIA demonstrated a strong association with both Aβ and tau pathology as indexed by PET. Endo-lysosomal proteins (e.g., HEXB, TPP1, SIAE) increased early in abundance alongside neurodegeneration-related proteins, and were followed by increases in metabolic proteins such as ALDOA, MDH1, and GOT1 at the mild cognitive impairment (MCI) stage. Finally, later AD stages were characterized by decreases in synaptic/membrane proteins (e.g., NPTX2). CONCLUSIONS Our study identified proxies of Aβ and tau pathology, indexed by PET, (SMOC1, YWHAE, CNN3) and highlighted the dynamic fluctuations of the CSF proteome over the disease course, identifying candidate biomarkers for disease staging beyond Aβ and tau.
Collapse
Affiliation(s)
- Sophia Weiner
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden.
| | - Mathias Sauer
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
| | - Laia Montoliu-Gaya
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
| | - Andrea L Benedet
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
| | - Nicholas J Ashton
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
- Banner Alzheimer's Institute and University of Arizona, Phoenix, AZ, USA
| | - Fernando Gonzalez-Ortiz
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Lab, Institute of Neuroscience and Physiology, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Joel Simrén
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
| | - Nesrine Rahmouni
- McGill University Research Centre for Studies in Aging, Montreal, QC, Canada
| | - Cecile Tissot
- McGill University Research Centre for Studies in Aging, Montreal, QC, Canada
| | - Joseph Therriault
- McGill University Research Centre for Studies in Aging, Montreal, QC, Canada
| | - Stijn Servaes
- McGill University Research Centre for Studies in Aging, Montreal, QC, Canada
| | - Jenna Stevenson
- McGill University Research Centre for Studies in Aging, Montreal, QC, Canada
| | - Ville Leinonen
- Department of Neurosurgery, NeuroCenter, Kuopio University Hospital and Neurosurgery, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Tuomas Rauramaa
- Department of Pathology, Kuopio University Hospital and Institute of Clinical Medicine-Pathology, University of Eastern Finland, Kuopio, Finland
| | - Mikko Hiltunen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Lab, Institute of Neuroscience and Physiology, Sahlgrenska University Hospital, Mölndal, Sweden
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute On Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, People's Republic of China
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Lab, Institute of Neuroscience and Physiology, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute, UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Johan Gobom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Lab, Institute of Neuroscience and Physiology, Sahlgrenska University Hospital, Mölndal, Sweden
| |
Collapse
|
12
|
Wang Y, Wu Z, Zheng Y, Wang H, Cheng B, Xia J. Unraveling the genetic underpinnings of mitochondrial traits and associated circulating inflammatory proteins in Alzheimer's disease: Mitochondrial HtrA2-T cell CD5 negative axis. J Alzheimers Dis 2025; 105:505-518. [PMID: 40170213 DOI: 10.1177/13872877251329517] [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: 04/03/2025]
Abstract
BackgroundPrevious studies with limited sample sizes have indicated a link between mitochondrial traits, inflammatory proteins, and Alzheimer's disease. The exact causality and their mediation relationships remain unclear.ObjectiveOur study aimed to delve into the genetic underpinnings of mitochondrial function and circulating inflammatory proteins in the pathogenesis of Alzheimer's disease.MethodsWe leveraged aggregated data from the largest genome-wide association study, including 69 mitochondrial traits, 91 circulating inflammatory proteins, and Alzheimer's disease. Bidirectional mendelian randomization (MR) analyses were performed to investigate their primary causal relationships. Thereafter a two-step MR mediation analysis was utilized to clarify the modulating effects of inflammatory proteins on mitochondria and Alzheimer's disease.ResultsOur study identified mitochondrial phenylalanine-tRNA ligase and 4-hydroxy-2-oxoglutarate aldolase as risk factors for Alzheimer's disease, and serine protease HtrA2 and carbonic anhydrase 5A as protective factors against Alzheimer's disease. Four inflammatory proteins (T-cell surface glycoprotein CD5, C-X-C motif chemokine 11, TGF-α, and TNF-related apoptosis-inducing ligand) played protective roles against Alzheimer's disease. Axin-1 and IL-6 increased the risk of Alzheimer's disease. Furthermore, T-cell surface glycoprotein CD5 was found to be a significant mediator between mitochondrial serine protease HTRA2 and Alzheimer's disease with the two-step MR method, accounting for 10.83% of the total effect.ConclusionsOur study emphasized mitochondrial HtrA2-T cell CD5 as a negative axis in Alzheimer's disease, offering novel perspectives on its etiology, pathogenesis, and treatment.
Collapse
Affiliation(s)
- Yixi Wang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Zhuokai Wu
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yiheng Zheng
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Haimeng Wang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Bin Cheng
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Juan Xia
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
13
|
Saberian P, Darvishi A, Khezragha D, Forouzandegan M, Farhadieh ME, Taghizadeh Khorshidi S, Hatami Nejad M, Sedighi S, Barati R, Safavi SAR, Sadeghi M, Gulisashvili D, Mayeli M, Shakeri S. MicroRNA Signatures Predict Brain Amyloidosis and Neurodegeneration in Alzheimer's Disease: Insights from [ 18F] AV45 and FDG PET Imaging. Brain Behav 2025; 15:e70572. [PMID: 40418688 DOI: 10.1002/brb3.70572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 04/25/2025] [Accepted: 04/28/2025] [Indexed: 05/28/2025] Open
Abstract
PURPOSE Alzheimer's disease (AD) is a neurodegenerative disease primarily manifesting with cognitive decline. This study aimed to investigate the alterations in microRNAs in patients across AD continuum as potential biomarkers. METHOD Data were extracted from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database, including microRNA levels in the serum and cerebrospinal fluid (CSF) of patients across AD continuum. We analyzed the associations between microRNA levels and previously known AD biomarkers, such as amyloid beta (Aβ) accumulations in the brain and glucose reuptake values using positron emission tomography ([18F]AV45 and FDG PET, respectively). FINDINGS The study found a significant positive correlation between CSF levels of miR-210-3p and Aβ accumulations in the brain (B = 4.69). Conversely, miR-223-3p levels were significantly lower in APOE-ε4 carriers. Significant negative correlations were also observed between glucose reuptake and several miRNAs in the AD group. Specifically, plasma levels of let-7g-5p, mir-423-5p, and mir-660-5p were negatively associated with glucose reuptake in the brain. CONCLUSION Elevated levels of miR-210-3p correlate with Aβ accumulation, supporting previous findings of increased levels of certain microRNAs in patients with MCI and AD. Our findings highlight the potential of microRNAs as biomarkers of AD.
Collapse
Affiliation(s)
- Parsa Saberian
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Afra Darvishi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Moojan Forouzandegan
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad-Erfan Farhadieh
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Sciences and Technology, University of Isfahan, Isfahan, Iran
| | | | - Mohamad Hatami Nejad
- Department of Psychology, Faculty of Psychology and Educational Sciences, University of Tehran, Tehran, Iran
| | - Sara Sedighi
- Department of Psychology, Faculty of Psychology and Educational Sciences, University of Tehran, Tehran, Iran
| | - Reza Barati
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Ahmad Reza Safavi
- Poursina Clinical Research Development Unit, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Sadeghi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- School of Rehabilitation, Shiraz University of Medical Sciences, Shiraz, Iran
| | - David Gulisashvili
- Department of Diagnostic Radiology & Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Mahsa Mayeli
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shayan Shakeri
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
14
|
Hoost SS, Honig LS, Kang MS, Bahl A, Lee AJ, Sanchez D, Reyes-Dumeyer D, Lantigua RA, Dage JL, Brickman AM, Manly JJ, Mayeux R, Gu Y. Association of dietary fatty acids with longitudinal change in plasma-based biomarkers of Alzheimer's disease. J Prev Alzheimers Dis 2025; 12:100117. [PMID: 40107919 PMCID: PMC12094269 DOI: 10.1016/j.tjpad.2025.100117] [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: 10/22/2024] [Revised: 01/29/2025] [Accepted: 02/27/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Elevated intake of omega-3 polyunsaturated fatty acids is linked to a reduced risk of dementia in some prospective studies. However, few studies have examined the relationship between nutrient intake and plasma biomarkers of Alzheimer's disease. OBJECTIVES We explored whether omega-3, omega-6, and monounsaturated fat intakes were associated with changes in plasma biomarkers of Alzheimer's disease over time. DESIGN The Washington Heights-Inwood Columbia Aging Project is a prospective cohort study (1994-2021); the data set used here includes a mean follow-up of 7.0 years. SETTING Community-based in New York City. PARTICIPANTS 599 dementia-free individuals at baseline who completed a 61-item food frequency questionnaire and had biomarkers measured in plasma from at least two different time points. MEASUREMENTS Fatty acid intake tertiles were computed from participant-completed 61-item Willett semi-quantitative food frequency questionnaires (Channing Laboratory, Cambridge, Massachusetts) obtained once at their baseline visit. Plasma-based biomarker assays were performed, using the single molecule array technology Quanterix Simoa HD-X platform, at baseline and follow-up visits. Generalized Estimating Equations (GEE) models were used to evaluate the association between baseline nutrient intake tertile and changes in biomarkers including phospho-tau181, amyloid-beta 42/40 ratio, phospho-tau181/amyloid-beta42 ratio, glial fibrillary acidic protein, neurofilament light chain, and two biomarker patterns derived from Principal Component Analysis (PCA1 and PCA2), with higher scores indicating a high level of neurodegeneration and low level of Alzheimer's disease burden, respectively). Models were adjusted for age, sex, race/ethnicity, education, and calculated total energy intake initially, and additionally for cerebrovascular risk factors. RESULTS Higher baseline omega-3 intake tertile was associated with lesser decline in PCA2 (β = 0.221, p < 0.001) and amyloid-beta 42/40 ratio (β = 0.022, p = 0.003), and a lesser rise in phospho-tau181 (β = -0.037, p = 0.001). Higher omega-6 intake tertile was linked to a lesser rise in phospho-tau181 (β = -0.050, p < 0.001) and glial fibrillary acidic protein (β = -0.028, p = 0.002). Most associations persisted after adjusting for cardiovascular risk factors. CONCLUSIONS Higher relative baseline intake of omega-3 and omega-6 fatty acids is associated with lesser progression of blood-based biomarkers of Alzheimer's disease. Consuming healthy fatty acids may help prevent accumulation of Alzheimer's disease-related pathological changes.
Collapse
Affiliation(s)
- Serena S Hoost
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, 710 West 168th Street, New York, New York, 10032, USA
| | - Lawrence S Honig
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA; G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, New York, 10032, USA; Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, 710 West 168th Street, New York, New York, 10032, USA
| | - Min Suk Kang
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA; Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, 710 West 168th Street, New York, New York, 10032, USA
| | - Aanya Bahl
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
| | - Annie J Lee
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA; Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, 710 West 168th Street, New York, New York, 10032, USA
| | - Danurys Sanchez
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA; Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, 710 West 168th Street, New York, New York, 10032, USA
| | - Dolly Reyes-Dumeyer
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA; Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, 710 West 168th Street, New York, New York, 10032, USA
| | - Rafael A Lantigua
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA; Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, 630 West 168th Street, New York, New York, 10032, USA
| | - Jeffrey L Dage
- Stark Neurosciences Research Institute, Suite 414, Indiana University School of Medicine, 320 West 15th Street, Indianapolis, Indiana, 46202, USA
| | - Adam M Brickman
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA; G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, New York, 10032, USA; Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, 710 West 168th Street, New York, New York, 10032, USA
| | - Jennifer J Manly
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA; G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, New York, 10032, USA; Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, 710 West 168th Street, New York, New York, 10032, USA
| | - Richard Mayeux
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA; G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, New York, 10032, USA; Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, 710 West 168th Street, New York, New York, 10032, USA; Department of Epidemiology, Mailman School of Public Health, Columbia University, 722 West 168th Street, New York, New York, 10032, USA
| | - Yian Gu
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY, 10032, USA; G.H. Sergievsky Center, Vagelos College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, New York, 10032, USA; Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, and the New York Presbyterian Hospital, 710 West 168th Street, New York, New York, 10032, USA; Department of Epidemiology, Mailman School of Public Health, Columbia University, 722 West 168th Street, New York, New York, 10032, USA.
| |
Collapse
|
15
|
Cohen AD, Villemagne VL. A brief history of Aβ imaging. Alzheimers Dement 2025; 21:e70291. [PMID: 40407091 PMCID: PMC12100503 DOI: 10.1002/alz.70291] [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: 04/03/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/26/2025]
Abstract
β-Amyloid (Αβ) imaging revolutionized the in vivo assessment of Alzheimer's disease (AD) Αβ pathology and its changes over time, increasing our insights into Aβ deposition in the brain by providing highly accurate, reliable, and reproducible quantitative statements of regional and global Aβ burden in the brain, proving essential for the differential diagnosis, staging, and evaluation of disease-specific anti-Αβ therapeutic approaches. Longitudinal observations, coupled with different disease-specific biomarkers to assess potential downstream effects of Aβ, have confirmed that Αβ deposition in the brain starts decades before the onset of symptoms. Aβ imaging studies continue to refine our understanding of the role of Αβ deposition in AD, and its relation to other imaging and fluid biomarkers. HIGHLIGHTS: Αβ imaging revolutionized the in vivo assessment of Alzheimer's disease Αβ pathology. Αβ imaging has increased our insights into Aβ deposition in the brain by providing highly accurate, reliable, and reproducible quantitative statements of regional and global Αβ burden in the brain. Αβ imaging is essential for the differential diagnosis, staging, and evaluation of disease-specific anti-Αβ therapeutic approaches. Αβ imaging studies continue to refine our understanding of the role of Αβ deposition in Alzheimer's disease, and its relation to other imaging and fluid biomarkers.
Collapse
Affiliation(s)
- Ann D. Cohen
- Department of PsychiatryThe University of PittsburghPittsburghPennsylvaniaUSA
| | | |
Collapse
|
16
|
Pan W, Teng Y, Han X, Liu S, Pang X, Wang L, Zhao M. Value of blood neural cell-derived small extracellular vesicles in the diagnosis and prediction of Alzheimer's disease: A systematic revie. J Prev Alzheimers Dis 2025:100193. [PMID: 40316481 DOI: 10.1016/j.tjpad.2025.100193] [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: 01/28/2025] [Revised: 04/17/2025] [Accepted: 04/19/2025] [Indexed: 05/04/2025]
Abstract
Blood neural cell-derived small extracellular vesicles (sEVs) can directly reflect changes in brain tissue and are easier to obtain than cerebrospinal fluid. This article systematically reviews the alterations of proteins and miRNAs from neural cell-derived sEVs in patients with Alzheimer's disease (AD), and summarizes the biomarkers with clinical diagnostic and predictive value. PubMed, Web of Science, Embase, and Cochrane Library were searched for studies in blood neural cell-derived sEVs in AD patients up to May 2024. According to the inclusion and exclusion criteria, the literature was screened, the information was extracted and the quality was evaluated. Proteins and miRNAs from neural cell-derived sEVs were classified and summarized, focusing on target molecules with high diagnostic and predictive values for AD. A final 34 articles reporting 5601 participants were included. In cross-sectional studies, Aβ- and Tau-related proteins (Aβ42, Aβ42/40, p-S396-Tau, p-Tau181), p-S312-IRS-1, and cathepsin D were increased, conversely, synaptic proteins (neurogranin, synaptotagmin, synaptophysin, synaptopodin, NMDAR2A) and REST were decreased in blood neuron-derived sEVs (NDsEVs) of patients with AD. While miR-29c-3p was increased in blood NDsEVs and glial cell-derived sEVs. Each of these proteins and miRNAs demonstrated high AD diagnostic value. Additionally, blood astrocyte-derived sEVs (ADsEVs) showed increased complement effector proteins and decreased complement regulatory proteins with a moderate diagnostic value. In longitudinal cohort studies, three composite models displayed high predictive efficacy for early AD prediction, and could predict the occurrence of AD within 1-10 years. Therefore, Aβ- and Tau-related proteins, synaptic proteins, and miRNA in blood neural cell-derived sEVs demonstrate high AD diagnostic and predictive values serving as important biomarkers. Especially, synaptic proteins showed significant changes in the early clinical stage, which has early predictive value.
Collapse
Affiliation(s)
- Weibing Pan
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Teng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaowan Han
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Shaojiao Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xingxue Pang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Lei Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| | - Mingjing Zhao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
17
|
Wei S, Li C, Li W, Yuan F, Kong J, Su X, Huang P, Guo H, Xu J, Sun H. Glial changes and gene expression in Alzheimer's disease from snRNA-Seq and spatial transcriptomics. J Alzheimers Dis 2025; 105:646-665. [PMID: 40267277 DOI: 10.1177/13872877251330320] [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: 04/25/2025]
Abstract
BackgroundAlzheimer's disease (AD) is characterized by cortical atrophy, glutamatergic neuron loss, and cognitive decline. However, large-scale quantitative assessments of cellular changes during AD pathology remain scarce.ObjectiveThis study aims to integrate single-nuclei sequencing data from the Seattle Alzheimer's Disease Cortical Atlas (SEA-AD) with spatial transcriptomics to quantify cellular changes in the prefrontal cortex and temporal gyrus, regions vulnerable to AD neuropathological changes (ADNC).MethodsWe mapped differentially expressed genes (DEGs) and analyzed their interactions with pathological factors such as APOE expression and Lewy bodies. Cellular proportions were assessed, focusing on neurons, glial cells, and immune cells.ResultsRORB-expressing L4-like neurons, though vulnerable to ADNC, exhibited stable cell numbers throughout disease progression. In contrast, astrocytes displayed increased reactivity, with upregulated cytokine signaling and oxidative stress responses, suggesting a role in neuroinflammation. A reduction in synaptic maintenance pathways indicated a decline in astrocytic support functions. Microglia showed heightened immune surveillance and phagocytic activity, indicating their role in maintaining cortical homeostasis.ConclusionsThe study underscores the critical roles of glial cells, particularly astrocytes and microglia, in AD progression. These findings contribute to a better understanding of cellular dynamics and may inform therapeutic strategies targeting glial cell function in AD.
Collapse
Affiliation(s)
- Songren Wei
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Institute for Brain Science and Intelligence, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China
| | - Chenyang Li
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Institute for Brain Science and Intelligence, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | | | - Fumiao Yuan
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jingjing Kong
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xi Su
- Women and Children Medical Research Center, Affiliated Foshan Women and Children Hospital, Foshan, China
| | - Peng Huang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- Women and Children Medical Research Center, Affiliated Foshan Women and Children Hospital, Foshan, China
| | - Hongbo Guo
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Institute for Brain Science and Intelligence, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jiangping Xu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China
| | - Haitao Sun
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Institute for Brain Science and Intelligence, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
18
|
Noda K, Kageyama I, Kobayashi Y, Lim Y, Sengoku S, Kodama K. Leveraging mHealth wearables for managing patients with Alzheimer's disease: a scoping review. Drug Discov Today 2025; 30:104363. [PMID: 40250750 DOI: 10.1016/j.drudis.2025.104363] [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: 04/04/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025]
Abstract
In this scoping review, we examine the role of wearable devices in diagnosing, treating, and monitoring Alzheimer's disease (AD) and mild cognitive impairment (MCI). It identifies various devices, including fitness trackers, smartwatches, electroencephalographic equipment, and sensors, which are used for monitoring physical activity, sleep patterns, and cognitive functions. Our review highlights the potential of these devices for early diagnosis and treatment, improving patient autonomy and quality of life. However, challenges, such as data privacy, device adherence, and technical limitations, remain. Future research should focus on integrating wearable devices with advanced diagnostic tools and validating their effectiveness across diverse populations.
Collapse
Affiliation(s)
- Kenta Noda
- Graduate School of Design and Architecture, Nagoya City University, Nagoya 464-0083, Japan; School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-Ku, Tokyo 142-8501, Japan
| | - Itsuki Kageyama
- School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-Ku, Tokyo 142-8501, Japan
| | - Yoshiyuki Kobayashi
- School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-Ku, Tokyo 142-8501, Japan
| | | | - Shintaro Sengoku
- School of Environment and Society, Institute of Science Tokyo, Tokyo 108-0023, Japan
| | - Kota Kodama
- School of Pharmacy and Pharmaceutical Sciences, Hoshi University, 2-4-41 Ebara, Shinagawa-Ku, Tokyo 142-8501, Japan; Ritsumeikan University, Osaka 567-8570, Japan; Center for Research and Education on Drug Discovery, The Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan.
| |
Collapse
|
19
|
Kolinger GD, Sotolongo-Grau O, Roé-Vellvé N, Tartari JP, Sanabria Á, Pérez-Martínez E, Koglin N, Stephens AW, Alegret M, Tárraga L, Gurruchaga MJ, Ruiz A, Boada M, Bullich S, Marquié M. Quantification of baseline amyloid PET in individuals with subjective cognitive decline can identify risk of amyloid accumulation and cognitive worsening: the FACEHBI study. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07270-7. [PMID: 40263206 DOI: 10.1007/s00259-025-07270-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 04/03/2025] [Indexed: 04/24/2025]
Abstract
PURPOSE Amyloid PET imaging is capable of measuring brain amyloid load in vivo. The aim of this study is to assess the relationship of the baseline amyloid with its accumulation over time and with cognition in individuals with subjective cognitive decline (SCD), giving a focus on those below Aβ positivity thresholds. METHODS 118 of 197 individuals with SCD from the Fundació ACE Healthy Brain Initiative underwent three [18F]florbetaben scans and the remaining 79 underwent two scans in a 5-year span. Individuals were categorised based on baseline Centiloid values (CL) into amyloid positive (Aβ+; CL > 35.7), Grey Zone (GZ; 20 < CL ≤ 35.7), and amyloid negative (Aβ-; CL ≤ 20). Relationship between conversion to mild cognitive decline (MCI) and baseline amyloid levels was assessed. Then, to focus on sub-threshold individuals with amyloid accumulation, the Aβ- group was split into two groups (N1 (CL ≤ 13.5) and N2 (13.5 < CL ≤ 20)), Aβ accumulation was determined, and a parametric image analysis of the Aβ accumulators in the N1 group was performed. RESULTS At baseline, 20 individuals were Aβ+, 8 GZ, 160 N1, and 9 N2. Higher Aβ load, older and less educated individuals presented increased risk of MCI-conversion. Longitudinally, 19% of N1 individuals were accumulators despite very low Aβ burden at baseline. Meanwhile, 89% of the N2 group accumulated Aβ as well as all GZ individuals (which had the highest rate of amyloid accumulation, 5.1 CL/year). In the parametric image analysis of N1 accumulators, a region within the precuneus was linked to increased Aβ over time. CONCLUSION Baseline amyloid levels differentiate individuals who accumulate amyloid over time and that are at risk for cognitive decline, including those at sub-threshold levels of Aβ. This can be valuable to identify pre-clinical AD in a SCD population.
Collapse
Grants
- 115952 Innovative Medicines Initiative
- 115975 Innovative Medicines Initiative
- 115985 Innovative Medicines Initiative
- PI13/02434 Spanish ISCIII, Acción Estratégica en Salud, integrated in the Spanish National R+D+I Plan and financed by ISCIII Subdirección General de Evaluación and the Fondo Europeo de Desarrollo Regional (FEDER "Una manera de hacer Europa")
- PI16/01861 Spanish ISCIII, Acción Estratégica en Salud, integrated in the Spanish National R+D+I Plan and financed by ISCIII Subdirección General de Evaluación and the Fondo Europeo de Desarrollo Regional (FEDER "Una manera de hacer Europa")
- PI19/01240 Spanish ISCIII, Acción Estratégica en Salud, integrated in the Spanish National R+D+I Plan and financed by ISCIII Subdirección General de Evaluación and the Fondo Europeo de Desarrollo Regional (FEDER "Una manera de hacer Europa")
- PI19/01301 Spanish ISCIII, Acción Estratégica en Salud, integrated in the Spanish National R+D+I Plan and financed by ISCIII Subdirección General de Evaluación and the Fondo Europeo de Desarrollo Regional (FEDER "Una manera de hacer Europa")
- PI22/00258 Spanish ISCIII, Acción Estratégica en Salud, integrated in the Spanish National R+D+I Plan and financed by ISCIII Subdirección General de Evaluación and the Fondo Europeo de Desarrollo Regional (FEDER "Una manera de hacer Europa")
- PI22/01403 Spanish ISCIII, Acción Estratégica en Salud, integrated in the Spanish National R+D+I Plan and financed by ISCIII Subdirección General de Evaluación and the Fondo Europeo de Desarrollo Regional (FEDER "Una manera de hacer Europa")
- PMP22/00022 European Union (NextGenerationEU)
- CB06/05/2004 CIBERNED (ISCIII)
- CB18/05/00010 CIBERNED (ISCIII)
- AC19/00097 Joint program for neurodegenerative diseases (JPND)
- PR067/21 Agency for Innovation and Entrepreneurship
- PI17/01474 ISCIII Subdirección General de Evaluación and the Fondo Europeo de Desarrollo Regional
- TARTAGLIA Programa Misiones de I+D en Inteligencia Artificial de la Secretaría de Estado de Digitalización e Inteligencia Artificial (SEDIA) del Ministerio de Asuntos Económicos y Transformación Digital
- 796706 HORIZON EUROPE Marie Sklodowska-Curie Actions
- PI19/00335 Subdirección General de Evaluación and the Fondo Europeo de Desarrollo Regional
Collapse
Affiliation(s)
| | - Oscar Sotolongo-Grau
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
| | | | - Juan Pablo Tartari
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
| | - Ángela Sanabria
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | | | | | | | - Montserrat Alegret
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Lluís Tárraga
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Miren Jone Gurruchaga
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
| | - Agustín Ruiz
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Mercè Boada
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Marta Marquié
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
20
|
Johnson KJ, Johnson K, Grant A, Taglialatela G, Micci MA. Photobiomodulation therapy increases neural stem cell pool in aged 3xTg-AD mice. PLoS One 2025; 20:e0321668. [PMID: 40261888 PMCID: PMC12013953 DOI: 10.1371/journal.pone.0321668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/10/2025] [Indexed: 04/24/2025] Open
Abstract
Presently approved Alzheimer's Disease (AD) therapeutics are designed for targeted removal of the AD-related toxic protein aggregate amyloid-β (Aβ) and have only shown moderate efficacy at slowing disease progression. Reversal of cognitive decline requires both removal of toxic aggregates and repair of the cellular systems damaged by decades of exposure to these aggregates. Adult hippocampal neurogenesis (AHN) is one such system that is known to be affected early and severely in the development of AD. Moreover, preserved AHN is associated with cognitive resilience to AD neuropathology. Therefore, targeted therapies to improve or enhance neurogenesis should be considered in addition to the removal of toxic protein aggregates. Photobiomodulation (PBM) using 670 nm LED light has been shown to induce synaptic resilience to and removal of AD-related toxic protein aggregates. In this study, we aimed to assess the effect of PBM on a mouse model of advanced AD neuropathology. Transgenic 3xTg-AD mice (15- to 17-month old) were randomized to receive PBM or SHAM therapy for one month, followed by neuropathological assessments. Our results show that one month of PBM therapy reduces hyperphosphorylated tau burden and partially rescues AHN in aged 3xTg-AD mice as compared to SHAM-treated transgenic mice. These data support the notion that PBM has the potential to be an effective non-invasive therapy to help preserve AHN and reduce cognitive dysfunction in moderate to advanced AD.
Collapse
Affiliation(s)
- Kevin J. Johnson
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Neurobiology, Neuroscience Graduate Program, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Kathia Johnson
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Auston Grant
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Giulio Taglialatela
- The Mitchell Center for Neurodegenerative Disorders, Department of Neurology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Maria-Adelaide Micci
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, United States of America
| |
Collapse
|
21
|
Feunaing RT, Gbaweng Yaya JA, Nyemb JN, Bassigue NI, Ketsemen HL, Henoumont C, Kandeda AK, Venditti A, Laurent S, Talla E. Antiradical and anti-acethylcholinesterase constituents from the methylene chloride extract of Ganoderma applanatum (Pers.) Pat (Ganodermataceae) and molecular docking study. Nat Prod Res 2025:1-13. [PMID: 40255072 DOI: 10.1080/14786419.2025.2491113] [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: 02/12/2025] [Accepted: 04/06/2025] [Indexed: 04/22/2025]
Abstract
Alzheimer's disease (AD) is a non-communicable disease with global impact. Inhibitors of acetylcholinesterase (AChE) are suitable therapies for AD. In this work, we report the isolation of antiacetylcholinesterase compounds from the methylene chloride (DCM) extract of the medical fungus Ganoderma applanatum (Pers.) Pat (Ganodermataceae). Chemical evaluation of this extract using chromatographic technics led to the isolation of a (1:1) mixture of ergosterol (1) and stellasterol (2), palmitic acid (3), ganodermanondiol (4), lucidumol B (5) and lupeol (6). Structures of these compounds were determined using spectroscopic analysis such as IR, MS, 1D & 2D NMR and literature. The acetylation reaction has been performed on the mixture (1 + 2) and compound 4, leading to the obtention the mixture of 3-acetyl-ergosterol (7) and 3-acetylstellasterol (8) along with 24-acetyl-ganodermanondiol (9) respectively. Total phenolic content was determined for DCM, Ethyl acetate and n-butanol extracts. To assess their antiradical scavenging potential, DPPH was used as free radical. The Inhibition power of acetylcholinesterase was evaluated in vitro using the Ellman reagent. Amongst all tested extracts, the DCM extract showed the high amount of total phenolic compounds with a value of 133.9512 mg EAG/g EX. The same extract showed a very good antiradical scavenging potential with an IC50 of 0.0021 mg/mL. The mixture (1 + 2) showed the highest antiradical scavenging activity with IC50 of 0.0770 mg/mL. The results obtained demonstrated that the acetylation has reduced the antiradical scavenging potential. Concerning the acetylcholinesterase inhibition power, the DCM extract and the mixture (1 + 2) showed a very good power with an inhibition percentage of 89%. The acetylation has also reduced the activity of the obtained derivative. The results provide insights into the potential efficacy of these compounds as acetylcholinesterase inhibitors. The binding interactions of the isolated and acetylated derivatives against acetylcholinesterase protein (PBP 3i6m) of Torpedo californica were studied using Autodock software. Ergosterol (-11.9 kcal/mol) binds better to the protein biding site through significant pi-sigma interactions.
Collapse
Affiliation(s)
- Roméo Toko Feunaing
- Department of Chemistry, Faculty of Science, University of Ngaoundere, Ngaoundere, Cameroon
| | - Joël Abel Gbaweng Yaya
- Department of Chemistry, Faculty of Science, University of Ngaoundere, Ngaoundere, Cameroon
- Centre for Research on Medicinal Plants and Traditional Medicine, Institute of Medical Research and Medicinal Plants Studies, Yaounde, Cameroon
| | - Jean Noël Nyemb
- Department of Refining and Petrochemistry, National Advanced School of Mines and Petroleum Industries, The University of Maroua, Kaele, Cameroon
| | - Noël Issa Bassigue
- Department of Chemistry, Faculty of Science, University of Ngaoundere, Ngaoundere, Cameroon
| | - Hervé Landry Ketsemen
- Department of Organic Chemistry, Faculty of Science, The University of Yaounde 1, Yaounde, Cameroon
| | - Céline Henoumont
- Department of General Organic and Biomedical Chemistry, Faculty of Medicine and Pharmacy, University of Mons, Belgium, Avenue Maistriau, Mons, Belgium
| | - Antoine Kavaye Kandeda
- Department of Biology and Animal Physiology, Faculty of Science, The University of Yaounde 1, Yaounde, Cameroon
| | | | - Sophie Laurent
- Department of Organic Chemistry, Faculty of Science, The University of Yaounde 1, Yaounde, Cameroon
| | - Emmanuel Talla
- Department of Chemistry, Faculty of Science, University of Ngaoundere, Ngaoundere, Cameroon
| |
Collapse
|
22
|
Décarie-Labbé L, Mellah S, Dialahy IZ, Belleville S. Predicting cognitive change using functional, structural, and neuropsychological predictors. Brain Commun 2025; 7:fcaf155. [PMID: 40337465 PMCID: PMC12056721 DOI: 10.1093/braincomms/fcaf155] [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: 03/31/2025] [Accepted: 04/17/2025] [Indexed: 05/09/2025] Open
Abstract
To effectively address Alzheimer's disease, it is crucial to understand its earliest manifestations, underlying mechanisms and early markers of progression. Recent findings of very early brain activation anomalies highlight their potential for early disease characterization and predicting future cognitive decline. Our objective was to evaluate the value of brain activation-both individually and in combination with structural and neuropsychological measures-for predicting cognitive change. The study included 105 individuals from the Consortium for the Early Identification of Alzheimer's Disease-Quebec cohort who exhibited subjective cognitive decline or mild cognitive impairment. Cognitive decline was assessed by calculating the slope of Montreal Cognitive Assessment scores using regression models across successive assessments, and individuals were characterized as either decliners or stable based on clinically reliable change. We evaluated cognitive decline predictions using unimodal models for each class of predictors and multimodal models that combined these predictors. Functional activation emerged as a strong predictor of cognitive change (R²=52.5%), with 87.6% accuracy and 98.7% specificity, performing comparably to structural and neuropsychological measures. Although the unimodal functional model exhibited high specificity, indicating that functional abnormalities frequently predict future decline, it had low sensitivity (60%), meaning that the absence of abnormalities does not rule out future decline. Multimodal models provided greater explanatory power than unimodal models and greater sensitivity than the functional model. These findings highlight the potential role of early brain activation anomalies in the early detection of future cognitive changes, offering valuable insights for clinicians and researchers in assessing cognitive decline risk and refining clinical trial criteria.
Collapse
Affiliation(s)
- Laurie Décarie-Labbé
- Research Center, Institut universitaire de gériatrie de Montréal, Montreal, Quebec, Canada, H3W 1W5
- Department of Psychology, Université de Montréal, Montreal, Quebec, Canada, H3C 3J7
| | - Samira Mellah
- Research Center, Institut universitaire de gériatrie de Montréal, Montreal, Quebec, Canada, H3W 1W5
| | - Isaora Z Dialahy
- Research Center, Institut universitaire de gériatrie de Montréal, Montreal, Quebec, Canada, H3W 1W5
| | - Sylvie Belleville
- Research Center, Institut universitaire de gériatrie de Montréal, Montreal, Quebec, Canada, H3W 1W5
- Department of Psychology, Université de Montréal, Montreal, Quebec, Canada, H3C 3J7
| |
Collapse
|
23
|
Akter S, Liu Z, Simoes EJ, Rao P. Using machine learning and electronic health record (EHR) data for the early prediction of Alzheimer's Disease and Related Dementias. J Prev Alzheimers Dis 2025:100169. [PMID: 40246680 DOI: 10.1016/j.tjpad.2025.100169] [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: 01/29/2025] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 04/19/2025]
Abstract
BACKGROUND Over 6 million patients in the United States are affected by Alzheimer's Disease and Related Dementias (ADRD). Early detection of ADRD can significantly improve patient outcomes through timely treatment. OBJECTIVE To develop and validate machine learning (ML) models for early ADRD diagnosis and prediction using de-identified EHR data from the University of Missouri (MU) Healthcare. DESIGN Retrospective case-control study. SETTING The study used de-identified EHR data provided by the MU NextGen Biomedical Informatics, modeled with the PCORnet Common Data Model (CDM). PARTICIPANTS An initial cohort of 380,269 patients aged 40 or older with at least two healthcare encounters was narrowed to a final dataset of 4,012 ADRD cases and 119,723 controls. METHODS Six ML classifier models: Gradient-Boosted Trees (GBT), Light Gradient-Boosting Machine (LightGBM), Random Forest (RF), eXtreme Gradient-Boosting (XGBoost), Logistic Regression (LR), and Adaptive Boosting (AdaBoost) were evaluated using Area Under the Receiver Operating Characteristic Curve (AUC-ROC), accuracy, sensitivity, specificity, and F1 score. SHAP (SHapley Additive exPlanations) analysis was applied to interpret predictions. RESULTS The GBT model achieved the best AUC-ROC scores of 0.809-0.833 across 1- to 5-year prediction windows. SHAP analysis identified depressive disorder, age groups 80-90 yrs and 70-80 yrs, heart disease, anxiety, and the novel risk factors of sleep apnea, and headache. CONCLUSION This study underscores the potential of ML models for leveraging EHR data to enable early ADRD prediction, supporting timely interventions, and improving patient outcomes. By identifying both established and novel risk factors, these findings offer new opportunities for personalized screening and management strategies, advancing both clinical and informatics science.
Collapse
Affiliation(s)
- Sonia Akter
- Institute for Data Science and Informatics, University of Missouri, USA
| | - Zhandi Liu
- Department of Electrical Engineering and Computer Science, University of Missouri, USA
| | - Eduardo J Simoes
- Department of Biomedical Informatics, Biostatics and Medical Epidemiology, University of Missouri, USA
| | - Praveen Rao
- Institute for Data Science and Informatics, University of Missouri, USA; Department of Electrical Engineering and Computer Science, University of Missouri, USA.
| |
Collapse
|
24
|
Bettcher BM, de Oliveira FF, Willette AA, Michalowska MM, Machado LS, Rajbanshi B, Borelli WV, Tansey MG, Rocha A, Suryadevara V, Hu WT. Analysis and interpretation of inflammatory fluid markers in Alzheimer's disease: a roadmap for standardization. J Neuroinflammation 2025; 22:105. [PMID: 40234920 PMCID: PMC11998147 DOI: 10.1186/s12974-025-03432-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 03/31/2025] [Indexed: 04/17/2025] Open
Abstract
Growing interest in the role of the immune response in Alzheimer's Disease and related dementias (ADRD) has led to widespread use of fluid inflammatory markers in research studies. To standardize the use and interpretation of inflammatory markers in AD research, we build upon prior guidelines to develop consensus statements and recommendations to advance application and interpretation of these markers. In this roadmap paper, we propose a glossary of terms related to the immune response in the context of biomarker discovery/validation, discuss current conceptualizations of inflammatory markers in research, and recommend best practices to address key knowledge gaps. We also provide consensus principles to summarize primary conceptual, methodological, and interpretative issues facing the field: (1) a single inflammatory marker is likely insufficient to describe an entire biological cascade, and multiple markers with similar or distinct functions should be simultaneously measured in a panel; (2) association studies in humans are insufficient to infer causal relationships or mechanisms; (3) neuroinflammation displays time-dependent and disease context-dependent patterns; (4) neuroinflammatory mechanisms should not be inferred based solely on blood inflammatory marker changes; and (5) standardized reporting of CSF inflammatory marker assay validation and performance will improve incorporation of inflammatory markers into the biological AD criteria.
Collapse
Affiliation(s)
- Brianne M Bettcher
- Department of Neurology, University of Colorado Anschutz Medical Campus, 12469 East 17th Place, Room 217- Campus Box F429, Aurora, CO, 80045, USA.
| | | | - Auriel A Willette
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School and Center for Healthy Aging Research, Rutgers Institute for Health, Health Care Policy, and Aging Research, Rutgers Health, New Brunswick, USA
| | - Malgorzata M Michalowska
- Department of Clinical Neuroscience, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Luiza Santos Machado
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Binita Rajbanshi
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California - San Francisco, San Francisco, USA
| | - Wyllians V Borelli
- Department of Morphological Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Malú Gámez Tansey
- Department of Neurology, Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, USA
| | - Andréia Rocha
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, USA
| | | | - William T Hu
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School and Center for Healthy Aging Research, Rutgers Institute for Health, Health Care Policy, and Aging Research, Rutgers Health, New Brunswick, USA
| |
Collapse
|
25
|
Wybitul M, Langer N, Hock C, Gietl A, Treyer V. Voxel-wise insights into early Alzheimer's disease pathology progression: the association with APOE and memory decline. GeroScience 2025:10.1007/s11357-025-01610-z. [PMID: 40167963 DOI: 10.1007/s11357-025-01610-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 03/08/2025] [Indexed: 04/02/2025] Open
Abstract
Longitudinal investigation of the Apolipoprotein E (APOE) genotype's impact on Alzheimer's disease (AD) biomarker progression, focusing on amyloid beta (Aβ) accumulation and gray matter (GM) atrophy, integrating cognitive decline and baseline levels. Longitudinal florbetapir-PET and T1-weighted MRI data from 100 cognitively normal (CN) and mild cognitive impaired (MCI) participants both with considerable global Aβ accumulation ("high Aβ accumulators") were analyzed using a voxel-wise approach. Associations of APOE genotype and memory decline with Aβ accumulation and GM atrophy were examined separately for each neuroimaging modality, controlling for baseline Aβ levels and diagnosis. Alternatively, the effect of baseline diagnosis, while controlling for memory decline, was investigated. A multimodal analysis evaluated interactions between genotype, memory decline, and GM atrophy on Aβ accumulation. High Aβ accumulators displayed extensive Aβ pathology predominantly in the medial orbito-frontal cortex, cingulate cortex, and precuneus, along with GM atrophy in temporal, occipital, orbito-frontal, and parietal areas. ɛ4 carriers with memory decline exhibited greater Aβ accumulation and GM atrophy in selective regions compared to non-carriers with memory decline, while no genotype difference was observed in individuals without decline. No interaction effect was observed for MCI diagnosis. Regional associations between the two biomarkers were similarly dependent on genotype and memory decline. ɛ4 carriers exhibiting memory decline present an accelerated neurobiological pattern at predementia stages, supporting early ɛ4 carrier monitoring and interventions in this at-risk group. Importantly, memory decline might be more informative than MCI regarding AD pathology progression emphasizing the importance of repeated cognitive assessments.
Collapse
Affiliation(s)
- Maha Wybitul
- Institute for Regenerative Medicine, Faculty of Medicine, University of Zurich, 8952, Schlieren, Switzerland
- Department of Psychology, Faculty of Philosophy, University of Zurich, 8050, Zurich, Switzerland
| | - Nicolas Langer
- Methods of Plasticity Research, Department of Psychology, University of Zurich, 8050, Zurich, Switzerland
| | - Christoph Hock
- Institute for Regenerative Medicine, Faculty of Medicine, University of Zurich, 8952, Schlieren, Switzerland
- Neurimmune, 8952, Schlieren, Switzerland
| | - Anton Gietl
- Institute for Regenerative Medicine, Faculty of Medicine, University of Zurich, 8952, Schlieren, Switzerland
- University Hospital of Psychiatry Zurich, Geriatric Psychiatry and Psychotherapy, 8008, Zurich, Switzerland
| | - Valerie Treyer
- Institute for Regenerative Medicine, Faculty of Medicine, University of Zurich, 8952, Schlieren, Switzerland.
- Department of Nuclear Medicine, University of Zurich, 8091, Zurich, Switzerland.
| |
Collapse
|
26
|
Wu Z, Long W, Yin Y, Tan B, Liu C, Li H, Ge S. Outer membrane vesicles of Porphyromonas gingivalis: recent advances in pathogenicity and associated mechanisms. Front Microbiol 2025; 16:1555868. [PMID: 40256625 PMCID: PMC12007433 DOI: 10.3389/fmicb.2025.1555868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 02/20/2025] [Indexed: 04/22/2025] Open
Abstract
Periodontitis is a chronic infectious inflammatory disease primarily caused by periodontal pathogenic bacteria, which poses a significant threat to human health. The pathogenic mechanisms associated with Porphyromonas gingivalis (P. gingivalis), a principal causative agent of periodontitis, are particularly complex and warrant thorough investigation. The extensive array of virulence factors released by this bacterium during its growth and pathogenesis not only inflicts localized damage to periodontal tissues but is also intricately linked to the development of systemic diseases through various mechanisms. The outer membrane vesicles (OMVs) produced by P. gingivalis play a key role in this process. These OMVs serve as important mediators of communication between bacteria and host cells and other bacteria, carrying and delivering virulence factors to host cells and distant tissues, thereby damaging host cells and exacerbating inflammatory responses. The ability of these OMVs to disseminate and deliver bacterial virulence factors allows P. gingivalis to play a pathogenic role far beyond the confines of the periodontal tissue and has been closely associated with the development of a variety of systemic diseases such as cardiovascular disease, Alzheimer's disease, rheumatoid arthritis, diabetes mellitus, non-alcoholic hepatitis, and cancer. In view of this, it is of great pathophysiological and clinical significance to deeply investigate its pathogenic role and related mechanisms. This will not only help to better understand the pathogenesis of periodontitis and its related systemic diseases but also provide new ideas and more effective and precise strategies for the early diagnosis, prevention, and treatment of these diseases. However, the current research in this field is still insufficient and in-depth, and many key issues and mechanisms need to be further elucidated. This article summarizes the recent research progress on the role of P. gingivalis OMVs (P. g-OMVs) in related diseases, with the aim of providing a theoretical basis and direction for future research and revealing the pathogenic mechanism of P. g-OMVs more comprehensively.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Song Ge
- School and Hospital of Stomatology, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
27
|
Aliaga A, Therriault J, Quispialaya K, Aliaga A, Kunach P, Macedo AC, Hopewell R, Rahmouni N, Soucy JP, Massarweh G, Guiot MC, Chan T, Klostranec J, Abreu Diaz AM, Rocha A, Carello-Collar G, Machado LS, De Bastiani MA, Guerini de Souza D, Souza DO, Zimmer AR, Gauthier S, Pascoal TA, Zimmer ER, Rosa-Neto P. Autoradiographic comparison between [ 11C]PiB and [ 18F]AZD4694 in human brain tissue. EJNMMI Res 2025; 15:30. [PMID: 40167827 PMCID: PMC11961831 DOI: 10.1186/s13550-025-01216-8] [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: 11/19/2024] [Accepted: 03/02/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Amyloid-β imaging through positron emission tomography (PET) has significantly transformed Alzheimer's disease (AD) research. [11C]PiB has been widely used for imaging β-amyloid plaques due to its high affinity and selectivity for amyloid deposits. [18F]AZD4694 is a more recently developed amyloid-PET imaging agent, which structurally resembles PiB and has less non-specific binding in the white matter than other 18F-labeled compounds. The purpose of this study is to compare the in vitro binding properties of the amyloid-PET radiotracers [11C]PiB and [18F]AZD4694 in post-mortem human brain tissue. Total binding was assessed by autoradiography in prefrontal, inferior parietal, posterior cingulate cortices and hippocampal sections of healthy control (HC) and AD autopsy-confirmed brain tissues. Furthermore, the displacement of [18F]AZD4694 by unlabeled PiB was evaluated in the above-mentioned sections of AD brain tissues. RESULTS For both radiotracers, we found significant differences (p < 0.0001) between HC and AD tissues binding in the prefrontal cortex ([11C]PiB Cohen's d = 3.424, [18F]AZD4694 Cohen's d = 5.070), inferior parietal cortex ([11C]PiB Cohen's d = 3.156, [18F]AZD4694 Cohen's d = 3.959), posterior cingulate cortex ([11C]PiB Cohen's d = 1.781, [18F]AZD4694 Cohen's d = 3.434), and hippocampus ([11C]PiB Cohen's d = 1.320, [18F]AZD4694 Cohen's d = 3.696). Higher binding was detected for [18F]AZD4694 compared to [11C]PiB in AD prefrontal, inferior parietal and posterior cingulate cortices, while binding in the hippocampus was comparable for both radioligands. Strong correlations between [18]AZD4694 and [11C]PiB were found in the prefrontal (R = 0.959, p < 0.0001), inferior parietal (R = 0.893, p < 0.0001), posterior cingulate (R = 0.838, p = 0.0006) cortices and hippocampus (R = 0.750, p < 0.0001). Bland-Altman analyses revealed strong agreement between [11C]PiB and [18F]AZD4694 in the prefrontal, inferior parietal, and posterior cingulate cortices, but lower agreement in the hippocampus. Displacement studies confirmed high binding affinity of PiB in all tissues, indicating that both amyloid-PET agents compete for the same binding sites. CONCLUSIONS This head-to-head study provides evidence that while [18F]AZD4694 and [11C]PiB bindings are highly correlated with both tracers competing for the same binding sites, [18F]AZD4694 has a slightly higher effect size when comparing between neuropathologically-confirmed AD and HC brain tissues.
Collapse
Affiliation(s)
- Antonio Aliaga
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC Room 3149, Montreal, QC, H4H 1R3, Canada
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal Do Rio Grande Do Sul, 2600 Ramiro Barcelos Street, Porto Alegre, Brazil
- Research Institute of the McGill University Health Centre, 1001, boul. Decarie -Bloc E, Offices ES2.1602, Montreal, QC, 1001H4A 3J1, Canada
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC Room 3149, Montreal, QC, H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
- Montreal Neurological Institute, Montreal, Canada
| | - Kely Quispialaya
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC Room 3149, Montreal, QC, H4H 1R3, Canada
- Research Institute of the McGill University Health Centre, 1001, boul. Decarie -Bloc E, Offices ES2.1602, Montreal, QC, 1001H4A 3J1, Canada
- Montreal Neurological Institute, Montreal, Canada
- Department of Experimental Medicine, McGill University, Montreal, Canada
| | - Arturo Aliaga
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC Room 3149, Montreal, QC, H4H 1R3, Canada
- Montreal Neurological Institute, Montreal, Canada
| | - Peter Kunach
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC Room 3149, Montreal, QC, H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
- Montreal Neurological Institute, Montreal, Canada
| | - Arthur C Macedo
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC Room 3149, Montreal, QC, H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
- Montreal Neurological Institute, Montreal, Canada
| | | | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC Room 3149, Montreal, QC, H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
- Montreal Neurological Institute, Montreal, Canada
| | | | | | - Marie-Christine Guiot
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
- Department of Pathology, McGill University Health Center, Montreal, Canada
| | - Tevy Chan
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC Room 3149, Montreal, QC, H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
- Montreal Neurological Institute, Montreal, Canada
| | - Jesse Klostranec
- Department of Diagnostic Radiology, McGill University Health Center, Montreal, Canada
| | - Aida Mary Abreu Diaz
- Department of Pharmacology and Physiology, University of Montreal, Montreal, Canada
| | - Andreia Rocha
- Department of Psychiatry, Pittsburgh University, Pittsburgh, USA
| | - Giovanna Carello-Collar
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal Do Rio Grande Do Sul, 2600 Ramiro Barcelos Street, Porto Alegre, Brazil
| | - Luiza S Machado
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal Do Rio Grande Do Sul, 2600 Ramiro Barcelos Street, Porto Alegre, Brazil
| | - Marco Antônio De Bastiani
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal Do Rio Grande Do Sul, 2600 Ramiro Barcelos Street, Porto Alegre, Brazil
| | - Débora Guerini de Souza
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal Do Rio Grande Do Sul, 2600 Ramiro Barcelos Street, Porto Alegre, Brazil
| | - Diogo O Souza
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal Do Rio Grande Do Sul, 2600 Ramiro Barcelos Street, Porto Alegre, Brazil
| | - Aline R Zimmer
- Department of Pharmacology, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC Room 3149, Montreal, QC, H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada
| | | | - Eduardo R Zimmer
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC Room 3149, Montreal, QC, H4H 1R3, Canada.
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal Do Rio Grande Do Sul, 2600 Ramiro Barcelos Street, Porto Alegre, Brazil.
- Graduate Program in Biological Sciences: Pharmacology and Therapeutics, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil.
- Brain Institute of Rio Grande Do Sul, Universidade Federal Do Rio Grande Do Sul, Porto Alegre, Brazil.
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, Douglas Hospital, McGill University, 6875 La Salle Blvd - FBC Room 3149, Montreal, QC, H4H 1R3, Canada.
- Research Institute of the McGill University Health Centre, 1001, boul. Decarie -Bloc E, Offices ES2.1602, Montreal, QC, 1001H4A 3J1, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montreal, Canada.
- Montreal Neurological Institute, Montreal, Canada.
- Department of Experimental Medicine, McGill University, Montreal, Canada.
| |
Collapse
|
28
|
Mackin RS, Rhodes E, Kassel M, Kryza-Lacombe M, Burns E, Bickford D, Morin R, Tosun D, Landau S, Butters MA, Aisen P, Raman R, Saykin AJ, Toga A, Koeppe R, Jack C, Weiner MW, Nelson C, Insel PS. Cortico-limbic volume abnormalities in late life depression are distinct from β amyloid and white matter pathologies. Mol Psychiatry 2025; 30:1267-1276. [PMID: 39511448 DOI: 10.1038/s41380-024-02677-4] [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: 10/27/2023] [Revised: 05/31/2024] [Accepted: 07/15/2024] [Indexed: 11/15/2024]
Abstract
This study was conducted to clarify patterns of cortico-limbic volume abnormalities in late life depression (LLD) relative to non-depressed (ND) adults matched for amyloid β (Aβ) deposition and to evaluate the relationship of volume abnormalities with cognitive performance. Participants included 116 LLD and 226 ND. Classification accuracy of LLD status was estimated using area under the receiver operator characteristic curve. Twenty-one percent of LLD and ND participants were Aβ positive and the groups did not differ on white matter hyperintensity volume (WMH (logscale); β = 0.12, p = 0.28). Compared to ND, the LLD group exhibited significantly lower bilateral volume in the lateral orbitofrontal cortex, hippocampus, accumbens area, superior temporal lobe, temporal pole, and amygdala after multiple comparison correction (p < 0.009 for all). Cortico-limbic volumes significantly improved classification of LLD beyond demographic characteristics, Aβ status, and WMH (AUCVol = 0.71, AUCWMH, Aβ = 0.62, AUC difference, 0.09 [0.03 to 0.15]). LLD exhibited poorer performance on measures of global cognition, set shifting, and verbal learning and memory relative to ND. Cognitive function was positively associated with cortico-limbic volumes and these relationships did not differ by group. Secondary analyses with an ND sample additionally matched for Mild Cognitive Impairment (MCI) diagnosis showed a similar but attenuated pattern of volume abnormalities. Overall, our results support LLD as being associated with cortico-limbic volume abnormalities that are distinct from Aβ and white matter pathologies and that these volume abnormalities are important factors associated with cognitive dysfunction in LLD.
Collapse
Affiliation(s)
- R Scott Mackin
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA.
- Veterans Administration Medical Center, San Francisco, CA, USA.
| | - Emma Rhodes
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
- Mental Illness Research Education and Clinical Centers, Veterans Administration Medical Center, San Francisco, CA, USA
| | - Michelle Kassel
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
- Mental Illness Research Education and Clinical Centers, Veterans Administration Medical Center, San Francisco, CA, USA
| | - Maria Kryza-Lacombe
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
- Mental Illness Research Education and Clinical Centers, Veterans Administration Medical Center, San Francisco, CA, USA
| | - Emily Burns
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
| | - David Bickford
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
| | - Ruth Morin
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
- Mental Illness Research Education and Clinical Centers, Veterans Administration Medical Center, San Francisco, CA, USA
| | - Duygu Tosun
- Veterans Administration Medical Center, San Francisco, CA, USA
- Department of Radiology, University of California, San Francisco, CA, USA
| | - Susan Landau
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| | - Meryl A Butters
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Paul Aisen
- University of Southern California, San Diego, CA, USA
- Alzheimer's Therapeutic Research Institute, San Diego, CA, USA
| | - Rema Raman
- University of Southern California, San Diego, CA, USA
- Alzheimer's Therapeutic Research Institute, San Diego, CA, USA
| | - Andrew J Saykin
- Indiana Alzheimer's Disease Research Center and the Department of Radiology and Imaging Sciences University School of Medicine, Indianapolis, IN, USA
| | - Arthur Toga
- Laboratory of Neuro Imaging, Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Robert Koeppe
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | | | - Michael W Weiner
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
- Veterans Administration Medical Center, San Francisco, CA, USA
- Department of Radiology, University of California, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Craig Nelson
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
| | - Philip S Insel
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
| |
Collapse
|
29
|
Andrey T, Alexander S, Inna B, Daria Z, Viktoria A, Anastasiia SG, Kumar A, Ivan F, Arina E, Oxana SG. Age as a limiting factor for effectiveness of photostimulation of brain drainage and cognitive functions. FRONTIERS OF OPTOELECTRONICS 2025; 18:6. [PMID: 40163163 PMCID: PMC11958890 DOI: 10.1007/s12200-025-00149-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 01/21/2025] [Indexed: 04/02/2025]
Abstract
The progressive number of old adults with cognitive impairment worldwide and the lack of effective pharmacologic therapies require the development of non-pharmacologic strategies. The photobiomodulation (PBM) is a promising method in prevention of early or mild age-related cognitive impairments. However, it remains unclear the efficacy of PBM for old patients with significant age-related cognitive dysfunction. In our study on male mice, we show a gradual increase in the brain amyloid beta (Aβ) levels and a decrease in brain drainage with age, which, however, is associated with a decline in cognitive function only in old (24 months of age) mice but not in middle-aged (12 months of age) and young (3 month of age) animals. These age-related features are accompanied by the development of hyperplasia of the meningeal lymphatic vessels (MLVs) in old mice underlying the decrease in brain drainage. PBM improves cognitive training exercises and Aβ clearance only in young and middle-aged mice, while old animals are not sensitive to PBM. These results clearly demonstrate that the PBM effects on cognitive function are correlated with age-mediated changes in the MLV network and may be effective if the MLV function is preserved. These findings expand fundamental knowledge about age differences in the effectiveness of PBM for improvement of cognitive functions and Aβ clearance as well as about the lymphatic mechanisms responsible for age decline in sensitivity to the therapeutic PBM effects.
Collapse
Affiliation(s)
- Terskov Andrey
- Department of Biology, Saratov State University, Saratov, 410012, Russia
| | - Shirokov Alexander
- Department of Biology, Saratov State University, Saratov, 410012, Russia
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Russian Academy of Sciences, Saratov, 410049, Russia
| | - Blokhina Inna
- Department of Biology, Saratov State University, Saratov, 410012, Russia
| | | | - Adushkina Viktoria
- Department of Biology, Saratov State University, Saratov, 410012, Russia
| | | | - Atul Kumar
- The Indian Institute of Technology (BHU) Varanasi, Uttar Pradesh, Varanasi, 221005, India
| | - Fedosov Ivan
- Institute of Physics, Saratov State University, Saratov, 410012, Russia
| | - Evsukova Arina
- Department of Biology, Saratov State University, Saratov, 410012, Russia
| | | |
Collapse
|
30
|
Modiri A, Abdolmaleki Z, Paryani MR. The effect of rosuvastatin coated by nano-chitosan on developing hippocampus: association with hippocampal neurogenesis and memory in an Alzheimer's induced model of rats. Anat Cell Biol 2025; 58:61-75. [PMID: 39914828 PMCID: PMC11933804 DOI: 10.5115/acb.24.250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/26/2024] [Accepted: 11/01/2024] [Indexed: 03/22/2025] Open
Abstract
Statins are long known to be beneficial for neurodegenerative conditions, including Alzheimer's disease (AD). Also, nanoparticle (NP) drugs can better affect the target tissue in various diseases. Therefore, the aim of this study was surveying the effect of rosuvastatin (RZV) coated by nano-chitosan in an Alzheimer's (Alz) induced model of rats. We examined learning, memory, and hippocampal amyloid plaques and evaluate expression levels of calbindin, doublecortin (DCX), NeuroD1, neuronal nuclei (NeuN), and neurofilament. Forty rats were randomly divided into five various groups. AD was induced by injecting bilaterally with 1 μl of amyloid beta (Aβ) into the hippocampus. After confirmation of AD, RZV, or NP, or RZV+NP were administered gavage orally daily in rats for 30 days. Induction of AD significantly raised Aβ plaques and dead cells compared to the control group. Results of Morris water maze in the test day indicated that Alz+NP+RZV group significantly reduced escape latency and travelled distance, also significantly increased spending time compared to the Alz group (P<0.05). RZV significantly decreased Aβ plaque percentage and the number of apoptotic cells compared to the Alz group (P<0.05). In addition, NeuN and neurofilament protein expression and calbindin, DCX, and NeuroD1 genes expression increased in Alz+RZV and Alz+RZV+NP compared to the Alz group. RZV coated by nano-chitosan has good potential for reducing Aβ plaques and dead cells, increasing brain NeuN and neurofilament proteins and calbindin, DCX, and NeuroD1 genes, and improving learning and memory in Alz rats.
Collapse
Affiliation(s)
- Armita Modiri
- Department of Pharmacology, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Zohreh Abdolmaleki
- Department of Pharmacology, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Mohammad Reza Paryani
- Department of Veterinary Basic Sciences, Karaj Branch, Islamic Azad University, Karaj, Iran
| |
Collapse
|
31
|
Lv H, Yi X, Guo X, Lin M, Bai D, Nie X, Wang X, Liu X. Correlation between mild cognitive impairment and flourishing among Chinese residents: a cross-sectional study. Front Psychol 2025; 16:1550013. [PMID: 40207108 PMCID: PMC11979250 DOI: 10.3389/fpsyg.2025.1550013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 03/12/2025] [Indexed: 04/11/2025] Open
Abstract
Objective This study aimed to investigate the correlation between mild cognitive impairment and flourishing among Chinese residents. Methods A total of 527 community residents aged ≥18 years were recruited from December 2023 to April 2024. Based on the results of the Ascertain Dementia 8-Item Informant Questionnaire (AD8), participants were classified into a healthy group (n = 356) and a mild cognitive impairment (MCI) group (n = 171). General demographic data, including age, gender, height, weight, place of residence, education level, marital status, household composition, personal income, occupation, and the flourishing scale (FS) were collected for statistical analysis. The analysis was performed using Statistical Product and Service Solutions software. Chi-square test was used to compare differences between the groups, while Kendall's correlation analysis and multivariate logistic regression were applied to assess the relationship between flourishing and MCI. Results Comparisons between the healthy and MCI groups showed that the FS scores in the healthy group were significantly higher than those in the MCI group (p < 0.01). Kendall's correlation analysis revealed that the score of AD8 was negatively correlated with FS (r = -0.237, p < 0.01). Multivariate analysis indicated that age [odds ratio (OR) = 1.451, 95% confidence interval (CI; 1.107-1.902), p = 0.007], place of residence [OR = 5.523, 95% CI (3.572-8.539), p < 0.001], and FS [OR = 0.421, 95%CI (0.311-0.569), p < 0.001] were correlated with MCI. Conclusion Flourishing levels are negatively correlated with MCI, and higher levels of flourishing associated with a lower risk of MCI. This suggests that flourishing may serve as a protective factor against cognitive decline. Additionally, age and place of residence are identified as risk factors for MCI.
Collapse
Affiliation(s)
- Hangqin Lv
- School of Nursing, Chengdu University, Chengdu, China
| | - Xin Yi
- School of Nursing, Chengdu University, Chengdu, China
| | - Xiangjun Guo
- School of Nursing, Chengdu University, Chengdu, China
| | - Meichuan Lin
- School of Nursing, Chengdu University, Chengdu, China
| | - Dingxi Bai
- School of Nursing, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | | | - Xue Wang
- School of Nursing, Chengdu University, Chengdu, China
| | - Xiaoyun Liu
- School of Nursing, Chengdu University, Chengdu, China
| |
Collapse
|
32
|
Jobin B, Magdamo C, Delphus D, Runde A, Reineke S, Soto AA, Ergun B, Mukhija S, Albers AD, Albers MW. The AROMHA brain health test is a remote olfactory assessment to screen for cognitive impairment. Sci Rep 2025; 15:9290. [PMID: 40128240 PMCID: PMC11933705 DOI: 10.1038/s41598-025-92826-8] [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: 07/31/2024] [Accepted: 03/03/2025] [Indexed: 03/26/2025] Open
Abstract
Cost-effective, noninvasive screening methods for preclinical Alzheimer's disease (AD) and other neurocognitive disorders remain an unmet need. The olfactory neural circuits develop AD pathological changes prior to symptom onset. To probe these vulnerable circuits, we developed the digital remote AROMHA Brain Health Test (ABHT), an at-home odor identification, discrimination, memory, and intensity assessment. The ABHT was self-administered among cognitively normal (CN) English and Spanish speakers (n = 127), participants with subjective cognitive complaints (SCC; n = 34), and mild cognitive impairment (MCI; n = 19). Self-administered tests took place remotely at home under unobserved (among interested CN participants) and observed modalities (CN, SCC, and MCI), as well as in-person with a research assistant present (CN, SCC, and MCI). Olfactory performance was similar across observed and unobserved remote self-administration and between English and Spanish speakers. Odor memory, identification, and discrimination scores decreased with age, and olfactory identification and discrimination were lower in the MCI group compared to CN and SCC groups, independent of age, sex, and education. The ABHT revealed age-related olfactory decline, and discriminated CN older adults from those with cognitive impairment. Replication of our results in other populations would support the use of the ABHT to identify and monitor individuals at risk for developing dementia.
Collapse
Affiliation(s)
- Benoît Jobin
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02129, USA
- Department of Psychology, Université du Québec à Trois-Rivières, Trois-Rivières, Canada
| | - Colin Magdamo
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02129, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Daniela Delphus
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02129, USA
| | - Andreas Runde
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02129, USA
| | | | | | - Beyzanur Ergun
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02129, USA
| | - Sasha Mukhija
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02129, USA
| | - Alefiya Dhilla Albers
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02129, USA.
- Department of Psychology, Endicott College, Beverly, MA, 01915, USA.
| | - Mark W Albers
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02129, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
- , 114 16th Street, Room 2003, Charlestown, MA, 02129, USA.
| |
Collapse
|
33
|
Cash DM, Morgan KE, O'Connor A, Veale TD, Malone IB, Poole T, Benzinger TL, Gordon BA, Ibanez L, Li Y, Llibre-Guerra JJ, McDade E, Wang G, Chhatwal JP, Day GS, Huey E, Jucker M, Levin J, Niimi Y, Noble JM, Roh JH, Sánchez-Valle R, Schofield PR, Bateman RJ, Frost C, Fox NC. Sample size estimates for biomarker-based outcome measures in clinical trials in autosomal dominant Alzheimer's disease. J Prev Alzheimers Dis 2025:100133. [PMID: 40118731 DOI: 10.1016/j.tjpad.2025.100133] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/07/2025] [Accepted: 03/09/2025] [Indexed: 03/23/2025]
Abstract
INTRODUCTION Alzheimer disease (AD)-modifying therapies are approved for treatment of early-symptomatic AD. Autosomal dominant AD (ADAD) provides a unique opportunity to test therapies in presymptomatic individuals. METHODS Using data from the Dominantly Inherited Alzheimer Network (DIAN), sample sizes for clinical trials were estimated for various cognitive, imaging, and CSF outcomes. Sample sizes were computed for detecting a reduction of either absolute levels of AD-related pathology (amyloid, tau) or change over time in neurodegeneration (atrophy, hypometabolism, cognitive change). RESULTS Biomarkers measuring amyloid and tau pathology had required sample sizes below 200 participants per arm (examples CSF Aβ42/40: 47[95 %CI 25,104], cortical PIB 49[28,99], CSF p-tau181 74[48,125]) for a four-year trial in presymptomatic individuals (CDR=0) to have 80 % power (5 % statistical significance) to detect a 25 % reduction in absolute levels of pathology, allowing 40 % dropout. For cognitive, MRI, and FDG, it was more appropriate to detect a 50 % reduction in rate of change. Sample sizes ranged from 250 to 900 (examples hippocampal volume: 338[131,2096], cognitive composite: 326[157,1074]). MRI, FDG and cognitive outcomes had lower sample sizes when including indivduals with mild impairment (CDR=0.5 and 1) as well as presymptomatic individuals (CDR=0). DISCUSSION Despite the rarity of ADAD, presymptomatic clinical trials with feasible sample sizes given the number of cases appear possible.
Collapse
Affiliation(s)
- David M Cash
- Dementia Research Centre, UCL Queen Square Institute of Neurology, First floor, 8-11 Queen Square, London, WC1N 3AR, UK; UK Dementia Research Institute, 6th Floor, Maple House, Tottenham Court Road, London W1T 7NF, UK.
| | - Katy E Morgan
- London School of Hygiene and Tropical Medicine, Keppel Street London, WC1E 7HT, UK
| | - Antoinette O'Connor
- Dementia Research Centre, UCL Queen Square Institute of Neurology, First floor, 8-11 Queen Square, London, WC1N 3AR, UK
| | - Thomas D Veale
- Dementia Research Centre, UCL Queen Square Institute of Neurology, First floor, 8-11 Queen Square, London, WC1N 3AR, UK
| | - Ian B Malone
- Dementia Research Centre, UCL Queen Square Institute of Neurology, First floor, 8-11 Queen Square, London, WC1N 3AR, UK
| | - Teresa Poole
- London School of Hygiene and Tropical Medicine, Keppel Street London, WC1E 7HT, UK
| | - Tammie Ls Benzinger
- Department of Radiology. Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110 USA
| | - Brian A Gordon
- Department of Radiology. Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110 USA; Knight Alzheimer Disease Research Center, Washington University School of Medicine, 4488 Forest Park Ave., Suite 200, St. Louis, MO 63108 USA
| | - Laura Ibanez
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA; Department of Psychiarty, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA
| | - Yan Li
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA
| | - Jorge J Llibre-Guerra
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA
| | - Eric McDade
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA
| | - Guoqiao Wang
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA
| | - Jasmeer P Chhatwal
- Brigham and Women's Hospital, Massachusetts General Hospital; Harvard Medical School, 75 Francis St, Boston, MA 02115, USA
| | - Gregory S Day
- Department of Neurology, Mayo Clinic, 4500 San Pablo Rd S, Jacksonville, FL 32224, USA
| | - Edward Huey
- Alpert Medical School of Brown University, Department of Psychiatry and Human Behavior, 222 Richmond St., Providence, RI 02903, USA
| | - Mathias Jucker
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Otfried-Müller Strasse 27, 72076 Tübingen, Germany; German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 23, 72076 Tübingen, Germany
| | - Johannes Levin
- Department of Neurology, LMU University Hospital, Marchioninistr. 15 D-81377, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17, 81377 Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Str. 17, 81377 Munich, Germany
| | - Yoshiki Niimi
- Unit for early and exploratory clinical development, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - James M Noble
- Department of Neurology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, GH Sergievksy Center, Columbia University, 710W 168th St #3, New York, NY 10032, USA
| | - Jee Hoon Roh
- Departments of Neurology and Physiology, Korea University Anam Hospital, Korea University College of Medicine, 73 goryeodae-ro, Seongbuk-gu, Seoul 02841, Republic Of Korea
| | - Racquel Sánchez-Valle
- Alzheimer's disease and other cognitive disorders group. Hospital Clínic de Barcelona. FRCB-IDIBAPS. University of Barcelona, Carrer de Villarroel, 170, L'Eixample, 08036 Barcelona, Spain
| | - Peter R Schofield
- Neuroscience Research Australia, Margarete Ainsworth Building Barker Street, Randwick NSW 2031 Australia; School of Biomedical Sciences, University of New South Wales, UNSW Sydney, NSW 2052 Australia
| | - Randall J Bateman
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, 4488 Forest Park Ave., Suite 200, St. Louis, MO 63108 USA; Department of Neurology, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA; Hope Center for Neurological Disorders, Washington University in St Louis, 4370 Duncan Ave., St. Louis, MO 63110, USA
| | - Chris Frost
- London School of Hygiene and Tropical Medicine, Keppel Street London, WC1E 7HT, UK
| | - Nick C Fox
- Dementia Research Centre, UCL Queen Square Institute of Neurology, First floor, 8-11 Queen Square, London, WC1N 3AR, UK; UK Dementia Research Institute, 6th Floor, Maple House, Tottenham Court Road, London W1T 7NF, UK
| |
Collapse
|
34
|
Liu H, Zhao X, Chen J, Win YY, Cai J. Unnatural foldamers as inhibitors of Aβ aggregation via stabilizing the Aβ helix. Chem Commun (Camb) 2025; 61:4586-4594. [PMID: 40035705 PMCID: PMC11878269 DOI: 10.1039/d4cc05280c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 02/24/2025] [Indexed: 03/06/2025]
Abstract
Protein aggregation is a critical factor in the development and progression of several human diseases, including Alzheimer's disease (AD), Huntington's disease, Parkinson's disease, and type 2 diabetes. Among these conditions, AD is recognized as the most prevalent progressive neurodegenerative disorder, characterized by the accumulation of amyloid-beta (Aβ) peptides. Neuronal toxicity is likely driven by soluble oligomeric intermediates of the Aβ peptide, which are thought to play a central role in the cascade leading to neuronal dysfunction and cognitive decline. In response, numerous therapeutic strategies have been developed to inhibit Aβ oligomerization, as this is believed to delay the formation of Aβ protofibrils. Traditional research has focused on discovering small molecules or peptides that antagonize Aβ oligomerization. However, recent studies have explored an alternative approach-developing ligands that stabilize the Aβ peptide in its α-helical conformation. This stabilization is thought to alter the peptide's natural aggregation kinetics, shifting it away from toxic oligomer formation and toward less harmful states. Crucially, by maintaining Aβ in this α-helical form, these ligands have been shown to rescue the peptide's associated cytotoxicity, offering a promising mechanism to mitigate the detrimental effects of Aβ in AD. While challenges remain, including treatment costs and side effects like ARIA (amyloid-related imaging abnormalities), anti-Aβ drug development represents a major advancement in Alzheimer's research and therapeutic options. This brief review aims to highlight the development and potential of these α-helix-stabilizing ligands as antagonists of Aβ aggregation, focusing on their interactions with Aβ and how these compounds induce and maintain secondary structural changes in the Aβ peptide. Notably, this innovative strategy holds promise beyond Aβ-related pathology, as the fundamental principles could be applied to other amyloidogenic proteins implicated in various amyloid-related diseases, potentially broadening the scope of therapeutic intervention for multiple neurodegenerative conditions.
Collapse
Affiliation(s)
- Heng Liu
- Department of Chemistry, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA.
| | - Xue Zhao
- Department of Chemistry, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA.
| | - Jianyu Chen
- Department of Chemistry, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA.
| | - Yu Yu Win
- Department of Chemistry, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA.
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA.
| |
Collapse
|
35
|
Heston MB, Teague JP, Cody KA, Deming Y, Ruiz de Chavez E, Morse J, Chin NA, Engelman CD, Chappell RJ, Langhough RE, Gleason CE, Clark LR, Zuelsdorff ML, Betthauser TJ, Alzheimer’s Disease Neuroimaging Initiative. Factors associated with age at tau pathology onset and time from tau onset to dementia. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.11.25323773. [PMID: 40162285 PMCID: PMC11952611 DOI: 10.1101/2025.03.11.25323773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
INTRODUCTION Elevated tau is temporally proximal to dementia onset but less is known about factors influencing T+ onset age and time to dementia following T+ in Alzheimer's disease. We used sampled iterative localized approximation (SILA) estimated T+ onset age (ETOA) to investigate factors associated with T+ age and time from T+ to dementia onset in ADNI. METHODS Using SILA-estimated A+ and T+ onset ages derived from 18F-Flortaucipir, 18F-Florbetapir, and 18F-Florbetaben PET and Cox proportional hazards and accelerated failure time models, we analyzed APOE, sex, amyloid burden, age, educational attainment, and literacy associations with ETOA and time from T+ to dementia. RESULTS Higher amyloid, APOE-ε4, lower education, and lower literacy associated with younger ETOA. Older ETOA and higher amyloid associated with shorter time from T+ to dementia. DISCUSSION This work highlights the prognostic value of ETOA and the need to better characterize factors contributing to ETOA and dementia onset in AD.
Collapse
Affiliation(s)
- Margo B. Heston
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, MC 2420, Madison, WI 53792-2420
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI 53705-2281
- Center for Health Disparities Research, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Madison, WI 53792-2420
| | - Jordan P. Teague
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, MC 2420, Madison, WI 53792-2420
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI 53705-2281
- Department of Medical Physics, University of Wisconsin-Madison School of Medicine and Public Health, 1111 Highland Ave, Rm 1005 Madison, WI 53705-2275
| | - Karly A. Cody
- Department of Neurology and Neurological Sciences, Stanford University Center for Academic Medicine, 453 Quarry Road, Palo Alto, CA 94304
| | - Yuetiva Deming
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, MC 2420, Madison, WI 53792-2420
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI 53705-2281
| | - Elena Ruiz de Chavez
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, MC 2420, Madison, WI 53792-2420
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI 53705-2281
| | - Jacob Morse
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, MC 2420, Madison, WI 53792-2420
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI 53705-2281
| | - Nathaniel A. Chin
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, MC 2420, Madison, WI 53792-2420
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI 53705-2281
| | - Corinne D. Engelman
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, MC 2420, Madison, WI 53792-2420
- Department of Population Health Sciences, University of Wisconsin-Madison School of Medicine and Public Health, 610 Walnut Street, Room 675, Madison, WI 53726
| | - Richard J. Chappell
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, MC 2420, Madison, WI 53792-2420
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison School of Medicine and Public Health, 610 Walnut Street, WARF Room 201, Madison, WI 53726
| | - Rebecca E. Langhough
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, MC 2420, Madison, WI 53792-2420
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI 53705-2281
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, 610 Walnut Street, 9th Floor, Madison, WI 53726
| | - Carey E. Gleason
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, MC 2420, Madison, WI 53792-2420
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI 53705-2281
- Madison Geriatric Research Education and Clinical Center (GRECC), William S. Middleton Veterans Affairs Hospital, 2500 Overlook Terrace, Madison, WI 53705
| | - Lindsay R. Clark
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, MC 2420, Madison, WI 53792-2420
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI 53705-2281
- Madison Geriatric Research Education and Clinical Center (GRECC), William S. Middleton Veterans Affairs Hospital, 2500 Overlook Terrace, Madison, WI 53705
| | - Megan L. Zuelsdorff
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, MC 2420, Madison, WI 53792-2420
- Center for Health Disparities Research, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Madison, WI 53792-2420
- University of Wisconsin-Madison School of Nursing, Signe Skott Cooper Hall, 701 Highland Avenue, Madison, WI 53705
| | - Tobey J. Betthauser
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, MC 2420, Madison, WI 53792-2420
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 1685 Highland Avenue, 5158 Medical Foundation Centennial Building, Madison, WI 53705-2281
- Center for Health Disparities Research, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Madison, WI 53792-2420
- Department of Medical Physics, University of Wisconsin-Madison School of Medicine and Public Health, 1111 Highland Ave, Rm 1005 Madison, WI 53705-2275
| | | |
Collapse
|
36
|
Nakaya M, Kamagata K, Takabayashi K, Andica C, Uchida W, Hagiwara A, Akashi T, Wada A, Taoka T, Naganawa S, Abe O, Aoki S. Magnetic resonance imaging indices for early Alzheimer's disease detection: Brain clearance markers. J Cereb Blood Flow Metab 2025:271678X251321305. [PMID: 40079562 PMCID: PMC11907505 DOI: 10.1177/0271678x251321305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
The Alzheimer's disease (AD) continuum is characterized by amyloid and tau protein deposition, which is partly attributable to the dysfunction of the brain clearance system. However, the specific phase in the AD continuum wherein aberrant clearance is present remains unclear. This study aimed to assess noninvasive magnetic resonance imaging (MRI) indices related to brain clearance functions, such as choroid plexus volume (CPV), lateral ventricular volume (LVV), and the index of diffusivity along the perivascular space (ALPS index), across the Alzheimer's disease (AD) spectrum. The CPV, LVV, and ALPS index in amyloid beta (Aβ)-negative healthy controls (HCs) and Aβ-positive HCs as well as in patients with Aβ-negative subjective cognitive decline (SCD), with Aβ-positive SCD, with mild cognitive impairment, and with AD were evaluated. The CPV and LVV were higher, whereas the ALPS index was lower in the patients with more severe disease. The ALPS index was significantly lower in Aβ-positive HCs than in Aβ-negative HCs. In SCD patients and those in the AD continuum, the MRI-based clearance markers were correlated with P-tau and T-tau protein levels and cognitive scores. In summary, brain clearance markers on MRI are associated with tau deposition, neurodegeneration and cognitive dysfunction.
Collapse
Affiliation(s)
- Moto Nakaya
- Department of Radiology, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Koji Kamagata
- Department of Radiology, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Kaito Takabayashi
- Department of Radiology, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Christina Andica
- Department of Radiology, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Faculty of Health Data Science, Juntendo University, Chiba, Japan
| | - Wataru Uchida
- Department of Radiology, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Akifumi Hagiwara
- Department of Radiology, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Toshiaki Akashi
- Department of Radiology, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Akihiko Wada
- Department of Radiology, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Toshiaki Taoka
- Department of Innovative Biomedical Visualization, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Shinji Naganawa
- Department of Radiology, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Osamu Abe
- Department of Radiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shigeki Aoki
- Department of Radiology, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Faculty of Health Data Science, Juntendo University, Chiba, Japan
| |
Collapse
|
37
|
Kumari A, Rahaman A, Zeng XA, Baloch Z. Therapeutic potential and microRNA regulating properties of phytochemicals in Alzheimer's disease. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102439. [PMID: 40114707 PMCID: PMC11925107 DOI: 10.1016/j.omtn.2024.102439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in the elderly and is characterized by the aggregation of Aβ (peptide) and neurofibrillary tangles along with inflammatory processes. Aging is a significant driver of these alterations, and dementia is a major cause of disability and mortality. Despite extensive clinical trials over the past two decades, no effective drug has been developed to improve AD symptoms or slow its progression, indicating the inefficiency of current treatment targets. In AD development, the molecular microenvironment plays a significant role. MicroRNAs (miRNAs) are a key component of this microenvironment, regulate post-transcriptional gene expression, and are expressed more abundantly in the brain than in other tissues. Several dysregulated miRNAs in AD have been linked to neuropathological changes, such as plaque and tangle accrual, as well as altered expression of notorious molecules. Preclinical studies have confirmed the efficacy of phytochemicals/food bioactive compounds (PCs/FBCs) in regulating miRNA expression, which makes them immensely beneficial for targeting miRNA-altered expression patterns in neuronal diseases. This review highlights the potential of miRNAs in driving AD pathology and its development. Furthermore, it discusses the therapeutic efficacy of PCs/FBCs and their miRNA-regulatory properties, especially focusing on antiinflammatory and antioxidant capacities for their development as effective AD agents.
Collapse
Affiliation(s)
- Ankita Kumari
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China
- Guangdong Key Laboratory of Food Intelligent Manufacturing, Foshan University, Foshan, Guangdong, China
- School of Food Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Abdul Rahaman
- Guangdong Key Laboratory of Food Intelligent Manufacturing, Foshan University, Foshan, Guangdong, China
- School of Food Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Xin-An Zeng
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China
- Guangdong Key Laboratory of Food Intelligent Manufacturing, Foshan University, Foshan, Guangdong, China
- School of Food Science and Engineering, Foshan University, Foshan, Guangdong, China
| | - Zulqarnain Baloch
- Faculty of Science and Technology, Kunming University of Science and Technology, Kunming, Yunan, China
| |
Collapse
|
38
|
Zeamer AL, Lai Y, Sanborn V, Loew E, Tracy M, Jo C, Ward DV, Bhattarai SK, Drake J, McCormick BA, Bucci V, Haran JP. Microbiome functional gene pathways predict cognitive performance in older adults with Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.06.641911. [PMID: 40161798 PMCID: PMC11952313 DOI: 10.1101/2025.03.06.641911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Disturbances in the gut microbiome is increasing correlated with neurodegenerative disorders, including Alzheimer's Disease. The microbiome may in fact influence disease pathology in AD by triggering or potentiating systemic and neuroinflammation, thereby driving disease pathology along the "microbiota-gut-brain-axis". Currently, drivers of cognitive decline and symptomatic progression in AD remain unknown and understudied. Changes in gut microbiome composition may offer clues to potential systemic physiologic and neuropathologic changes that contribute to cognitive decline. Here, we recruited a cohort of 260 older adults (age 60+) living in the community and followed them over time, tracking objective measures of cognition, clinical information, and gut microbiomes. Subjects were classified as healthy controls or as having mild cognitive impairment based on cognitive performance. Those with a diagnosis of Alzheimer's Diseases with confirmed using serum biomarkers. Using metagenomic sequencing, we found that relative species abundances correlated well with cognition status (MCI or AD). Furthermore, gene pathways analyses suggest certain microbial metabolic pathways to either be correlated with cognitive decline or maintaining cognitive function. Specifically, genes involved in the urea cycle or production of methionine and cysteine predicted worse cognitive performance. Our study suggests that gut microbiome composition may predict AD cognitive performance.
Collapse
Affiliation(s)
- Abigail L. Zeamer
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Yushuan Lai
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | | | - Ethan Loew
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Matthew Tracy
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Cynthia Jo
- Department of Emergency Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Doyle V. Ward
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Shakti K. Bhattarai
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | | | - Beth A. McCormick
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Vanni Bucci
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - John P. Haran
- Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Department of Emergency Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, University of Massachusetts Chan Medical School, Worcester, MA, USA
| |
Collapse
|
39
|
Wang K, Adjeroh DA, Fang W, Walter SM, Xiao D, Piamjariyakul U, Xu C. Comparison of Deep Learning and Traditional Machine Learning Models for Predicting Mild Cognitive Impairment Using Plasma Proteomic Biomarkers. Int J Mol Sci 2025; 26:2428. [PMID: 40141072 PMCID: PMC11941952 DOI: 10.3390/ijms26062428] [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: 01/16/2025] [Revised: 03/01/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
Mild cognitive impairment (MCI) is a clinical condition characterized by a decline in cognitive ability and progression of cognitive impairment. It is often considered a transitional stage between normal aging and Alzheimer's disease (AD). This study aimed to compare deep learning (DL) and traditional machine learning (ML) methods in predicting MCI using plasma proteomic biomarkers. A total of 239 adults were selected from the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort along with a pool of 146 plasma proteomic biomarkers. We evaluated seven traditional ML models (support vector machines (SVMs), logistic regression (LR), naïve Bayes (NB), random forest (RF), k-nearest neighbor (KNN), gradient boosting machine (GBM), and extreme gradient boosting (XGBoost)) and six variations of a deep neural network (DNN) model-the DL model in the H2O package. Least Absolute Shrinkage and Selection Operator (LASSO) selected 35 proteomic biomarkers from the pool. Based on grid search, the DNN model with an activation function of "Rectifier With Dropout" with 2 layers and 32 of 35 selected proteomic biomarkers revealed the best model with the highest accuracy of 0.995 and an F1 Score of 0.996, while among seven traditional ML methods, XGBoost was the best with an accuracy of 0.986 and an F1 Score of 0.985. Several biomarkers were correlated with the APOE-ε4 genotype, polygenic hazard score (PHS), and three clinical cerebrospinal fluid biomarkers (Aβ42, tTau, and pTau). Bioinformatics analysis using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) revealed several molecular functions and pathways associated with the selected biomarkers, including cytokine-cytokine receptor interaction, cholesterol metabolism, and regulation of lipid localization. The results showed that the DL model may represent a promising tool in the prediction of MCI. These plasma proteomic biomarkers may help with early diagnosis, prognostic risk stratification, and early treatment interventions for individuals at risk for MCI.
Collapse
Affiliation(s)
- Kesheng Wang
- Department of Biobehavioral Health & Nursing Science, College of Nursing, University of South Carolina, Columbia, SC 29208, USA
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Donald A. Adjeroh
- Lane Department of Computer Science & Electrical Engineering, West Virginia University, Morgantown, WV 26506, USA;
| | - Wei Fang
- West Virginia Clinical and Translational Science Institute, Morgantown, WV 26506, USA;
| | - Suzy M. Walter
- School of Nursing, Health Sciences Center, West Virginia University, Morgantown, WV 26506, USA; (S.M.W.); (U.P.)
| | - Danqing Xiao
- Department of STEM, School of Arts and Sciences, Regis College, Weston, MA 02493, USA;
| | - Ubolrat Piamjariyakul
- School of Nursing, Health Sciences Center, West Virginia University, Morgantown, WV 26506, USA; (S.M.W.); (U.P.)
| | - Chun Xu
- Department of Health and Biomedical Sciences, College of Health Professions, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| |
Collapse
|
40
|
Cash DM, Morgan KE, O’Connor A, Veale TD, Malone IB, Poole T, Benzinger TLS, Gordon BA, Ibanez L, Li Y, Llibre-Guerra JJ, McDade E, Wang G, Chhatwal JP, Day GS, Huey E, Jucker M, Levin J, Niimi Y, Noble JM, Roh JH, Sánchez-Valle R, Schofield PR, Bateman RJ, Frost C, Fox NC, The Dominantly Inherited Alzheimer Network (DIAN). Sample size estimates for biomarker-based outcome measures in clinical trials in autosomal dominant Alzheimer's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2024.11.12.24316919. [PMID: 39606328 PMCID: PMC11601746 DOI: 10.1101/2024.11.12.24316919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
INTRODUCTION Alzheimer disease (AD)-modifying therapies are approved for treatment of early-symptomatic AD. Autosomal dominant AD (ADAD) provides a unique opportunity to test therapies in presymptomatic individuals. METHODS Using data from the Dominantly Inherited Alzheimer Network (DIAN), sample sizes for clinical trials were estimated for various cognitive, imaging, and CSF outcomes. Sample sizes were computed for detecting a reduction of either absolute levels of AD-related pathology (amyloid, tau) or change over time in neurodegeneration (atrophy, hypometabolism, cognitive change). RESULTS Biomarkers measuring amyloid and tau pathology had required sample sizes below 200 participants per arm (examples CSF Aβ42/40: 47[95%CI 25,104], cortical PIB 49[28,99], CSF p-tau181 74[48,125]) for a four-year trial in presymptomatic individuals (CDR=0) to have 80% power (5% statistical significance) to detect a 25% reduction in absolute levels of pathology, allowing 40% dropout. For cognitive, MRI, and FDG, it was more appropriate to detect a 50% reduction in rate of change. Sample sizes ranged from 250-900 (examples hippocampal volume: 338[131,2096], cognitive composite: 326[157,1074]). MRI, FDG and cognitive outcomes had lower sample sizes when including indivduals with mild impairment (CDR=0.5 and 1) as well as presymptomatic individuals (CDR=0). DISCUSSION Despite the rarity of ADAD, presymptomatic clinical trials with feasible sample sizes given the number of cases appear possible.
Collapse
Affiliation(s)
- David M Cash
- Dementia Research Centre, UCL Queen Square Institute of Neurology, First floor, 8-11 Queen Square, London, WC1N 3AR, UK
- UK Dementia Research Institute, 6th Floor, Maple House, Tottenham Court Road, London W1T 7NF, UK
| | - Katy E Morgan
- London School of Hygiene and Tropical Medicine, Keppel Street London, WC1E 7HT, UK
| | - Antoinette O’Connor
- Dementia Research Centre, UCL Queen Square Institute of Neurology, First floor, 8-11 Queen Square, London, WC1N 3AR, UK
| | - Thomas D Veale
- Dementia Research Centre, UCL Queen Square Institute of Neurology, First floor, 8-11 Queen Square, London, WC1N 3AR, UK
| | - Ian B Malone
- Dementia Research Centre, UCL Queen Square Institute of Neurology, First floor, 8-11 Queen Square, London, WC1N 3AR, UK
| | - Teresa Poole
- London School of Hygiene and Tropical Medicine, Keppel Street London, WC1E 7HT, UK
| | - Tammie LS Benzinger
- Department of Radiology. Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110 USA
| | - Brian A Gordon
- Department of Radiology. Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110 USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, 4488 Forest Park Ave., Suite 200, St. Louis, MO 63108 USA
| | - Laura Ibanez
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA
- Department of Psychiarty, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA
| | - Yan Li
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA
| | - Jorge J. Llibre-Guerra
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA
| | - Eric McDade
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA
| | - Guoqiao Wang
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA
| | - Jasmeer P Chhatwal
- Brigham and Women’s Hospital; Massachusetts General Hospital; Harvard Medical School, 75 Francis St, Boston, MA 02115, USA
| | - Gregory S Day
- Department of Neurology, Mayo Clinic, 4500 San Pablo Rd S, Jacksonville, FL 32224, USA
| | - Edward Huey
- Alpert Medical School of Brown University, Department of Psychiatry and Human Behavior, 222 Richmond St., Providence, RI 02903, USA
| | - Mathias Jucker
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Hoppe-Seyler-Straße 3, 72076 Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Otfried-Müller-Straße 23, 72076 Tübingen, Germany
| | - Johannes Levin
- Department of Neurology, LMU University Hospital, Marchioninistr. 15 D-81377, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen-Strasse 17,81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Str. 17, 81377 Munich, Germany
| | - Yoshiki Niimi
- Unit for early and exploratory clinical development, The UniVersity of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - James M Noble
- Department of Neurology, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, GH Sergievksy Center, Columbia University, 710 W 168th St #3, New York, NY 10032, USA
| | - Jee Hoon Roh
- Departments of Neurology and Physiology, Korea University Anam Hospital, Korea University College of Medicine, 73 goryeodae-ro, Seongbuk-gu, Seoul 02841, Republic Of Korea
| | - Racquel Sánchez-Valle
- Alzheimer’s disease and other cognitive disorders group. Hospital Clínic de Barcelona. FRCB-IDIBAPS. University of Barcelona, Carrer de Villarroel, 170, L’Eixample, 08036 Barcelona, Spain
| | - Peter R Schofield
- Neuroscience Research Australia, Margarete Ainsworth Building Barker Street, Randwick NSW 2031 Australia
- School of Biomedical Sciences, University of New South Wales, UNSW Sydney, NSW 2052 Australia
| | - Randall J Bateman
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, 4488 Forest Park Ave., Suite 200, St. Louis, MO 63108 USA
- Department of Neurology, Washington University in St Louis, 660 S. Euclid Ave., St. Louis, MO 63110 USA
- Hope Center for Neurological Disorders, Washington University in St Louis, 4370 Duncan Ave., St. Louis, MO 63110, USA
| | - Chris Frost
- London School of Hygiene and Tropical Medicine, Keppel Street London, WC1E 7HT, UK
| | - Nick C Fox
- Dementia Research Centre, UCL Queen Square Institute of Neurology, First floor, 8-11 Queen Square, London, WC1N 3AR, UK
- UK Dementia Research Institute, 6th Floor, Maple House, Tottenham Court Road, London W1T 7NF, UK
| | | |
Collapse
|
41
|
Zhao J, Li Z, Zhang R, Yu H, Zhang L. Network pharmacology mechanism of Rosmarinus officinalis L.(Rosemary) to improve cell viability and reduces apoptosis in treating Alzheimer's disease. BMC Complement Med Ther 2025; 25:94. [PMID: 40055645 PMCID: PMC11889937 DOI: 10.1186/s12906-025-04771-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 01/20/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disorder characterized by neurodegeneration, nerve loss, neurofibrillary tangles and Aβ plaques. Different process of the AD pathology present more opportunities for treatment. In addition, the holistic approaches involving network pharmacology with traditional Chinese medicine (TCM) may be viable options for AD treatment, and lead to an effective cure for AD in the future. Therefore, this study explored the therapeutic effect and mechanism of Rosmarinus officinalis L(rosemary) in the treatment of Alzheimer's disease on basis of cell experiments, network pharmacology and molecular docking. METHODS We performed cell experiments, to investigate the therapeutic effects of Rosmarinus officinalis L on AD in vitro using CCK8 assay, flow cytometry and TMRE Kit. In addition, carnosic acid is a major antioxidant diterpenoid in Rosmarinus officinalis L. We performed cell experiments, to investigate the neuroprotective effects of carnosic acid on AD in virto using CCK8 assay and flow cytometry. Furthermore, the main antioxidant compounds of rosemary ware collected through literature reviews, PharmMapper and Swiss Target prediction ware used to identify their potential targets. Targets of AD were obtained from Genecards and OMIM. The intersection targets of the main active components of rosemary and the therapeutic targets of Alzheimer's disease were subsequently obtained by using online Venn diagram. Protein-protein interaction, Cytoscape, Gene Ontology, and Kyoto Encyclopedia of Genes were used to analyze potential targets and key pathways of rosemary in AD. Besides, through molecular docking, the interactions of the main active components of rosemary, and the predicted candidate targets were verified. Finally, quantitative Real-Time PCR (RT-qPCR) was performed to confirm the effectiveness of the genes. RESULTS It was found that rosemary could reversed Aβ25-35 induced damage to mouse hippocampal neuron HT22 cells, significantly improved the viability of damaged cells, and reduced apoptosis. The results of fluorescent staining with Hoechst 33,342 and TMRE suggested that rosemary inhibited the reduction of mitochondrial membrane potential levels induced by Aβ25-35, which had a specific protective effect on AD in vitro. Additionally, a main antioxidant compound in rosemary, carnosic acid, also has neuroprotective effects. Eight main antioxidant compounds of rosemary ware collected. Network pharmacology and molecular docking, revealed that rosemary plays a therapeutic role in the treatment of Alzheimer's disease through the main active carnosic acid, carnosol, rosmarinol, rosmadial, genkwanin, cirsimaritin, rosmarinic acid and caffeic acid in Rosmarinus officinalis L, which affects the gene regulation of HRAS, ESR1, RHOA, IGF1, SRC, ANXA5, MMP9, MAPK14, NOS3, and PIK3R1, and participates in the PI3K-Akt, Rap1, MAPK, and estrogen signaling pathways. RT-qPCR indicated that rosemary could elevated expression of IGF1, MMP9 and decreased mRNA levels of SRC, MAPK14, compared with the control group. CONCLUSIONS Rosemary is an important economic plant with multi-component, multi-target and multi-pathway synergistic effects.The findings highlight the effectiveness of rosemary in helping to increase cell viability and reduce apoptosis when treating mouse hippocampal neuron HT22 cells, thereby supporting its therapeutic potential in treating Alzheimer's disease.
Collapse
Affiliation(s)
- Jingzhi Zhao
- Department of Laboratory Animal Science, School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Zhejian Li
- Department of Laboratory Animal Science, School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Rongping Zhang
- Yunnan Key Laboratory of Southern Medicinal Resources, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan, 650500, China
| | - Haofei Yu
- Department of Laboratory Animal Science, School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, 650500, China.
| | - Lanchun Zhang
- Department of Laboratory Animal Science, School of Pharmaceutical Science & Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, 650500, China.
| |
Collapse
|
42
|
Cornea M, Vintilă BI, Bucuța M, Ștef L, Anghel CE, Grama AM, Lomnasan A, Stetiu AA, Boicean A, Sava M, Paziuc LC, Manea MC, Tîbîrnă A, Băcilă CI. Efficacy of Transcranial Direct Current Stimulation and Photobiomodulation in Improving Cognitive Abilities for Alzheimer's Disease: A Systematic Review. J Clin Med 2025; 14:1766. [PMID: 40095881 PMCID: PMC11900501 DOI: 10.3390/jcm14051766] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 02/24/2025] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
Background: Due to the increasing global prevalence of Alzheimer's dementia (AD), neuromodulation techniques such as transcranial direct current stimulation (tDCS) and photobiomodulation (PBM) are considered potential complementary therapies. Objective: We assessed the efficacy and safety of tDCS and PBM and their potential to enhance cognitive functions in individuals with AD. Methods: This review primarily examined studies designed to evaluate the efficacy, followed by an assessment of the safety of tDCS and PBM for people with AD. The databases searched were PubMed, Scopus, and Web of Science Core Collection, resulting in 17 published randomized and controlled trials. References were screened over 5 years (2020-2024). The research design used PRISMA guidelines. Results: Fourteen studies were considered for tDCS, and the current literature supports efficacy and safety at an amperage of 2 mA, with electrodes placed on the dorsolateral prefrontal cortex (DLPFC). Three studies were included for PBM. The heterogeneity of these study measures made them unsuitable for combined efficacy analysis, and they did not provide a safety evaluation. Conclusions: Despite differences in efficacy assessments, tDCS and PBM improved cognitive abilities. There is an urgent need to standardize metrics for evaluating efficacy and safety, particularly for PBM. Future research is encouraged.
Collapse
Affiliation(s)
- Monica Cornea
- “Dr. Gheorghe Preda” Clinical Psychiatry Hospital of Sibiu, 550082 Sibiu, Romania; (M.C.); (C.E.A.); (A.M.G.); (A.L.); (C.-I.B.)
| | - Bogdan Ioan Vintilă
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (L.Ș.); (A.A.S.); (A.B.); (M.S.)
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania
- Neuroscience Scientific Research Collective, 550082 Sibiu, Romania
| | - Mihaela Bucuța
- Faculty of Social Sciences and Humanities, Lucian Blaga University of Sibiu, 550024 Sibiu, Romania
| | - Laura Ștef
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (L.Ș.); (A.A.S.); (A.B.); (M.S.)
| | - Claudia Elena Anghel
- “Dr. Gheorghe Preda” Clinical Psychiatry Hospital of Sibiu, 550082 Sibiu, Romania; (M.C.); (C.E.A.); (A.M.G.); (A.L.); (C.-I.B.)
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (L.Ș.); (A.A.S.); (A.B.); (M.S.)
- Neuroscience Scientific Research Collective, 550082 Sibiu, Romania
| | - Andreea Maria Grama
- “Dr. Gheorghe Preda” Clinical Psychiatry Hospital of Sibiu, 550082 Sibiu, Romania; (M.C.); (C.E.A.); (A.M.G.); (A.L.); (C.-I.B.)
| | - Andrei Lomnasan
- “Dr. Gheorghe Preda” Clinical Psychiatry Hospital of Sibiu, 550082 Sibiu, Romania; (M.C.); (C.E.A.); (A.M.G.); (A.L.); (C.-I.B.)
| | - Andreea Angela Stetiu
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (L.Ș.); (A.A.S.); (A.B.); (M.S.)
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania
| | - Adrian Boicean
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (L.Ș.); (A.A.S.); (A.B.); (M.S.)
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania
| | - Mihai Sava
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (L.Ș.); (A.A.S.); (A.B.); (M.S.)
- County Clinical Emergency Hospital of Sibiu, 550245 Sibiu, Romania
| | - Lucian Constantin Paziuc
- Campulung Moldovenesc Psychiatric Hospital, Trandafirilor Street 2, 725100 Câmpulung Moldovenesc, Romania;
| | - Mihnea Costin Manea
- “Prof. Dr. Alexandru Obregia” Clinical Hospital of Psychiatry, 041914 Bucharest, Romania; (M.C.M.); (A.T.)
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Bvd, 050474 Bucharest, Romania
| | - Andrian Tîbîrnă
- “Prof. Dr. Alexandru Obregia” Clinical Hospital of Psychiatry, 041914 Bucharest, Romania; (M.C.M.); (A.T.)
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Bvd, 050474 Bucharest, Romania
| | - Ciprian-Ionuț Băcilă
- “Dr. Gheorghe Preda” Clinical Psychiatry Hospital of Sibiu, 550082 Sibiu, Romania; (M.C.); (C.E.A.); (A.M.G.); (A.L.); (C.-I.B.)
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania; (L.Ș.); (A.A.S.); (A.B.); (M.S.)
- Neuroscience Scientific Research Collective, 550082 Sibiu, Romania
| |
Collapse
|
43
|
B A, Kalirajan K. An intelligent magnetic resonance imagining-based multistage Alzheimer's disease classification using swish-convolutional neural networks. Med Biol Eng Comput 2025; 63:885-899. [PMID: 39546213 DOI: 10.1007/s11517-024-03237-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 10/28/2024] [Indexed: 11/17/2024]
Abstract
Alzheimer's disease (AD) refers to a neurological disorder that causes damage to brain cells and results in decreasing cognitive abilities and memory. In brain scans, this degeneration can be seen in different ways. The disease can be classified into four stages: Non-demented (ND), moderate demented (MoD), mild demented (MiD), and very mild demented (VMD). To prepare the raw dataset for analysis, the collected magnetic resonance imaging (MRI) images are subjected to several pre-processing techniques in order to improve the performance accuracy of the proposed model. Medical images generally have poor contrast and get affected by noise, which ends up with inaccurate diagnosis. For the different phases of AD to be detected, a clear image is necessary. To address this issue, the influence of the artefacts must be reduced, enhance the contrast, and reduce the loss of information. A novel framework for image enhancement is suggested to increase the accuracy in the detection and identification of AD. In this study, the raw MRI dataset from the Alzheimer's disease neuroimaging initiative (ADNI) database is subjected to skull stripping, contrast enhancement, and image filtering followed by data augmentation to balance the dataset with four types of Alzheimer's classes. The pre-processed data are subjected to five different pre-trained models such as AlexNet, ResNet, VGG 16, EfficientNet, and Inceptionv3 achieving a testing accuracy rate of 91.2%, 88.21%, 92.34%, 93.45%, and 85.12%, respectively. These pre-trained models are compared with the proposed CNN (convolutional neural network) model designed with Adam optimizer and Flatten Swish activation function which reaches the highest accuracy of 96.5% with a learning rate of 0.000001. The five pre-trained CNN models along with the proposed swish-based AD-CNN were tested using various performance metrics to evaluate the model efficiency in classifying and identifying the AD classes. From the result analysis, it is evident that the proposed AD-CNN model outperforms all the other models.
Collapse
Affiliation(s)
- Archana B
- Department of Electronics and Communication Engineering, KGiSL Institute of Technology, Coimbatore, Tamil Nadu, India.
| | - K Kalirajan
- Department of Electronics and Communication Engineering, KPR Institute of Engineering and Technology, Coimbatore, Tamil Nadu, India
| |
Collapse
|
44
|
Mao X, Li A, Wang Y, Wang Y, Ren S, He K, Guan Y, Huang Q, Guo Q, Li Z, Guo T, Xie F. Association of objective subtle cognitive difficulties with amyloid-β and tau deposition compared to subjective cognitive decline. Eur J Nucl Med Mol Imaging 2025; 52:1481-1495. [PMID: 39836215 DOI: 10.1007/s00259-025-07080-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
PURPOSE This study evaluated the differences in amyloid-β (Aβ), tau deposition, and longitudinal tau deposition between subjective cognitive decline (SCD) and objective subtle cognitive difficulties (Obj-SCD). METHODS Participants from the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort (n = 234) and the Huashan cohort (n = 267) included individuals with Obj-SCD, SCD, subjective memory concern (SMC), and healthy controls (HC). General linear models (GLM) were used to compare baseline and longitudinal differences in Aβ and tau among the groups, and to examine the associations between these biomarkers. RESULTS Obj-SCD participants had significantly higher Aβ and tau levels compared to HC, with increased annual accumulation of Aβ and tau, especially in Braak stages III-IV. In contrast, SCD/SMC participants did not show significant differences from HC in annual Aβ and tau changes. Baseline Aβ PET correlated with annual tau PET changes in Obj-SCD (Braak III-VI) and SCD/SMC (all Braak stages), and baseline Aβ levels strongly predicted early memory decline in both Obj-SCD and SCD/SMC groups. CONCLUSION This study indicates that Obj-SCD is more strongly associated with Aβ and tau biomarkers, with Aβ contributing to the progression of tau pathology and memory decline. Further research should include more longitudinal data to more robustly validate these findings and clarify the temporal relationship between Aβ and tau in preclinical Alzheimer's disease.
Collapse
Affiliation(s)
- Xiaoxie Mao
- School of Medicine, Xiamen University, 361102, Xiamen, Fujian, China
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, 361102, Xiamen, Fujian, China
| | - Anqi Li
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Ying Wang
- Department of Gerontology, Shanghai Jiaotong University Affiliated Sixth People's Hospital, 200233, Shanghai, China
| | - Yan Wang
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Shuhua Ren
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Kun He
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yihui Guan
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Qi Huang
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Qihao Guo
- Department of Gerontology, Shanghai Jiaotong University Affiliated Sixth People's Hospital, 200233, Shanghai, China
| | - Zijing Li
- School of Medicine, Xiamen University, 361102, Xiamen, Fujian, China.
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, 361102, Xiamen, Fujian, China.
| | - Tengfei Guo
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China.
| | - Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China.
- MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
45
|
Soncu Büyükişcan E. Neuropsychology of Alzheimer's disease: From preclinical phase to dementia. APPLIED NEUROPSYCHOLOGY. ADULT 2025:1-9. [PMID: 39982692 DOI: 10.1080/23279095.2025.2469236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by significant cognitive and functional decline, initially presenting with episodic memory impairment. A thorough neuropsychological assessment is essential for AD diagnosis, particularly in the early stages in which interventions may be more effective. This paper reviews the neuropsychology of Alzheimer's disease, highlighting the cognitive progression of the disease. In the typical forms of AD, episodic memory appears to be the first and foremost affected cognitive domain. As AD progresses, cognitive impairments extend beyond memory to affect various domains such as attention, executive functions, language, and visuospatial abilities. Neuropsychiatric issues, such as depression and anxiety, which often accompany cognitive decline, are also common, especially at the advanced stages of the disease. While episodic memory impairment is the earliest and most prominent feature in typical AD cases, comprehensive assessments, including social cognition and neuropsychiatric evaluations, are crucial for accurate diagnosis and treatment planning.
Collapse
|
46
|
Rosal AE, Martin SL, Strafella AP. The role of Apolipoprotein E4 on cognitive impairment in Parkinson's disease and Parkinsonisms. Front Neurosci 2025; 19:1515374. [PMID: 40052092 PMCID: PMC11882537 DOI: 10.3389/fnins.2025.1515374] [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: 10/22/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025] Open
Abstract
Cognitive impairment is a prevalent non-motor symptom of Parkinson's disease (PD), increasing the risk of dementia as the disease progresses. Despite its clinical significance, the etiology of cognitive impairment in PD remains unclear. Apolipoprotein E4 (APOE4), a well-known genetic risk factor of Alzheimer's disease, has been studied for its potential role in PD-related cognitive impairment. However, findings have been conflicting and thus inconclusive, highlighting a need to critically evaluate the current research. Several studies using neuroimaging modalities have explored the brains of individuals with PD and atypical parkinsonian disorders who have APOE4. Some of these studies have identified distinct neuropathological changes that have been previously reported to be associated with cognitive impairments in those with Parkinsonisms. Here, we review the role of APOE4 on cognitive impairment in PD and atypical Parkinsonisms using neuroimaging evidence. We will examine how APOE4 may contribute to pathological changes within the brain and its association with cognitive impairment.
Collapse
Affiliation(s)
- Angenelle Eve Rosal
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Institute of Medical Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Sarah L. Martin
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Translation and Computational Neurosciences Unit (TCNU), Faculty of Health and Education, Manchester Metropolitan University, Manchester, United Kingdom
| | - Antonio P. Strafella
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Institute of Medical Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Edmond J. Safra Parkinson Disease Program, Neurology Division, Toronto Western Hospital and Krembil Brain Institute, University Health Network, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
47
|
Zheng C, Zhao W, Yang Z, Guo S, the Alzheimer’s Disease Neuroimaging Initiative. Dysfunction in the hierarchy of morphometric similarity network in Alzheimer's disease and its correlation with cognitive performance and gene expression profiles. Psychol Med 2025; 55:e42. [PMID: 39934009 PMCID: PMC12055026 DOI: 10.1017/s0033291725000091] [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: 07/03/2024] [Revised: 12/05/2024] [Accepted: 01/02/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND Previous research has shown abnormal functional network gradients in Alzheimer's disease (AD). Structural network gradient is capable of capturing continuous changes in brain morphology and has the ability to elucidate the underlying processes of neurodevelopment. However, it remains unclear whether structural network gradients are altered in AD and what associations exist between these changes and cognitive function, and gene expression profiles. METHODS By constructing an individualized structural network gradient decomposition framework, we calculated the morphological similarity network (MSN) gradients for 404 subjects (186 AD patients and 218 normal controls). We investigated AD-related alterations in MSN gradients, along with the associations between MSN gradients and cognitive function, MSN topological properties, and gene expression profiles. RESULTS Our findings indicated that the principal MSN gradient alterations in AD were primarily characterized by an increase in the primary and secondary sensory cortices and a decrease in the association cortex 1. The primary and higher-order cortices exhibited opposite associations with cognition, including executive function, language skills, and memory processes. Moreover, the principal MSN gradients were found to significantly predict cognitive function in AD. The altered gradient pattern was 14.8% attributable to gene expression profiles, and the genes demonstrating the highest correlation are involved in metabolic activity and synaptic signaling. CONCLUSIONS Our results offered novel insights into the underlying mechanisms of structural brain network impairment in AD patients, enhancing our understanding of the neurobiological processes responsible for impaired cognition in patients with AD, and offering a new dimensional structural biomarker for AD.
Collapse
Affiliation(s)
- Chuchu Zheng
- School of Public Health, Shanxi Medical University, Taiyuan, People’s Republic of China
- MOE-LCSM, School of Mathematics and Statistics, Hunan Normal University, Changsha, People’s Republic of China
- Key Laboratory of Applied Statistics and Data Science, Hunan Normal University, College of Hunan Province, Changsha, People’s Republic of China
| | - Wei Zhao
- MOE-LCSM, School of Mathematics and Statistics, Hunan Normal University, Changsha, People’s Republic of China
- Key Laboratory of Applied Statistics and Data Science, Hunan Normal University, College of Hunan Province, Changsha, People’s Republic of China
| | - Zeyu Yang
- MOE-LCSM, School of Mathematics and Statistics, Hunan Normal University, Changsha, People’s Republic of China
- Key Laboratory of Applied Statistics and Data Science, Hunan Normal University, College of Hunan Province, Changsha, People’s Republic of China
| | - Shuixia Guo
- MOE-LCSM, School of Mathematics and Statistics, Hunan Normal University, Changsha, People’s Republic of China
- Key Laboratory of Applied Statistics and Data Science, Hunan Normal University, College of Hunan Province, Changsha, People’s Republic of China
| | | |
Collapse
|
48
|
Qu T. The effects of amyloidosis and aging on glutamatergic and GABAergic synapses, and interneurons in the barrel cortex and non-neocortical brain regions. Front Neuroanat 2025; 19:1526962. [PMID: 40012738 PMCID: PMC11863279 DOI: 10.3389/fnana.2025.1526962] [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: 11/12/2024] [Accepted: 01/15/2025] [Indexed: 02/28/2025] Open
Abstract
Previous studies on changes in the distribution of GABAergic interneurons and excitation/inhibition (E/I) balance in Alzheimer's disease (AD) and aging were mainly conducted in the neocortex and hippocampus. However, the limbic system is the primary and crucial location for AD progression. Therefore, in this study, we utilized AD and aging mouse models to investigate the E/I balance and the distribution of parvalbumin (PV)- and somatostatin (SST)-expressing cells in S1BF (barrel field of primary somatosensory cortex, barrel cortex), CA1 hippocampal area and brain regions beyond the neocortex and hippocampus, including retrosplenial cortex (RSC, which is composed of RSG and RSA), piriform cortex (Pir), amygdala (BMA), and hypothalamus (DM). We discovered that amyloidosis may disrupt the alignment of excitatory pre- and postsynaptic quantities. Amyloidosis reduces the quantity of synapses and SST cells, but does not impact the counts of PV cells. By contrast, aging is linked to a decline in synapses, I/E ratios, SST and PV cells. Amyloidosis affects the S1BF and BMA, while aging may harm all studied regions, including the S1BF, RSC, hippocampus, Pir, BMA, and DM. Aging mostly affects synapses and I/E ratios in Pir, BMA, and DM, and PV and SST interneurons in the hippocampus.
Collapse
Affiliation(s)
- Tao Qu
- Molecular Neuroplasticity, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
49
|
Hosseinpouri A, Sadegh K, Zarei-Behjani Z, Dehghan Z, Karbalaei R. Identification of critical genes and drug repurposing targets in entorhinal cortex of Alzheimer's disease. Neurogenetics 2025; 26:27. [PMID: 39928227 DOI: 10.1007/s10048-025-00806-x] [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: 11/18/2024] [Accepted: 01/31/2025] [Indexed: 02/11/2025]
Abstract
Alzheimer's disease (AD) is a slow brain degeneration disorder in which the accumulation of beta-amyloid precursor plaque and an intracellular neurofibrillary tangle of hyper-phosphorylated tau proteins in the brain have been implicated in neurodegeneration. In this study, we identified the most important genes that are unique and sensitive in the entorhinal region of the brain to target AD effectively. At first, microarrays data are selected and constructed protein-protein interaction network (PPIN) and gene regulatory network (GRN) from differentially expressed genes (DEGs) using Cytoscape software. Then, networks analysis was performed to determine hubs, bottlenecks, clusters, and signaling pathways in AD. Finally, critical genes were selected as targets for repurposing drugs. Analyzing the constructed PPIN and GRN identified CD44, ELF1, HSP90AB1, NOC4L, BYSL, RRP7A, SLC17A6, and RUVBL2 as critical genes that are dysregulated in the entorhinal region of AD suffering patients. The functional enrichment analysis revealed that DEG nodes are involved in the synaptic vesicle cycle, glutamatergic synapse, PI3K-Akt signaling pathway, retrograde endocannabinoid signaling, endocrine and other factor-regulated calcium reabsorption, ribosome biogenesis in eukaryotes, and nicotine addiction. Gentamicin, isoproterenol, and tumor necrosis factor are repurposing new drugs that target CD44, which plays an important role in the development of AD. Following our model validation using the existing experimental data, our model based on previous experimental reports suggested critical molecules and candidate drugs involved in AD for further investigations in vitro and in vivo.
Collapse
Affiliation(s)
- Arghavan Hosseinpouri
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Khadijeh Sadegh
- Department of Biological Science and Technology, Faculty of Nano and Bio Science and Technology, Persian Gulf University, Bushehr, Iran
| | - Zeinab Zarei-Behjani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zeinab Dehghan
- Department of Comparative Biomedical Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
- Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Reza Karbalaei
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, USA.
| |
Collapse
|
50
|
Fischer L, Molloy EN, Pichet Binette A, Vockert N, Marquardt J, Pacha Pilar A, Kreissl MC, Remz J, Tremblay-Mercier J, Poirier J, Rajah MN, Villeneuve S, Maass A. Precuneus Activity during Retrieval Is Positively Associated with Amyloid Burden in Cognitively Normal Older APOE4 Carriers. J Neurosci 2025; 45:e1408242024. [PMID: 39788739 PMCID: PMC11800745 DOI: 10.1523/jneurosci.1408-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 01/12/2025] Open
Abstract
The precuneus is a site of early amyloid-beta (Aβ) accumulation. Previous cross-sectional studies reported increased precuneus fMRI activity in older adults with mild cognitive deficits or elevated Aβ. However, longitudinal studies in early Alzheimer's disease (AD) are lacking and the relationship to the Apolipoprotein-E (APOE) genotype is unclear. Investigating the PREVENT-AD dataset, we assessed how baseline and longitudinal precuneus activity during successful memory retrieval relates to future Aβ and tau burden and change in memory performance. We further studied the moderation by APOE4 genotype. We included 165 older adults (age, 62.8 ± 4.4 years; 113 female; 66 APOE4 carriers) who were cognitively normal at baseline with a family history of AD. All participants performed task-fMRI at baseline and underwent 18F-flortaucipir-PET and 18F-NAV4694-Aβ-PET on average 5 years later. We found that higher baseline activity and greater longitudinal increase in precuneus activity were associated with higher Aβ burden in APOE4 carriers but not noncarriers. We observed no effects of precuneus activity on tau burden. Finally, APOE4 noncarriers with low baseline precuneus activity exhibited better longitudinal performance in an independent memory test compared with (1) noncarriers with higher baseline activity and (2) APOE4 carriers. Our findings suggest that higher task-related precuneus activity during memory retrieval at baseline and over time are associated with greater Aβ burden in cognitively normal APOE4 carriers. Our results further indicate that the absence of "hyperactivation" and the absence of the APOE4 allele is related with better future cognitive outcomes in cognitively normal older adults at risk for AD.
Collapse
Affiliation(s)
- Larissa Fischer
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg 39120, Germany
| | - Eóin N Molloy
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg 39120, Germany
- Division of Nuclear Medicine, Department of Radiology & Nuclear Medicine, Faculty of Medicine, Otto von Guericke University Magdeburg, Magdeburg 39120, Germany
| | - Alexa Pichet Binette
- Clinical Memory Research, Faculty of Medicine, Lund University, Lund 223 62, Sweden
- Douglas Mental Health University Institute Research Centre, McGill University, Montréal H4H 1R3, Canada
| | - Niklas Vockert
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg 39120, Germany
| | - Jonas Marquardt
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg 39120, Germany
| | - Andrea Pacha Pilar
- Institute for Biology, Otto von Guericke University Magdeburg, Magdeburg 39120, Germany
| | - Michael C Kreissl
- Division of Nuclear Medicine, Department of Radiology & Nuclear Medicine, Faculty of Medicine, Otto von Guericke University Magdeburg, Magdeburg 39120, Germany
| | - Jordana Remz
- Douglas Mental Health University Institute Research Centre, McGill University, Montréal H4H 1R3, Canada
| | - Jennifer Tremblay-Mercier
- Douglas Mental Health University Institute Research Centre, McGill University, Montréal H4H 1R3, Canada
| | - Judes Poirier
- Douglas Mental Health University Institute Research Centre, McGill University, Montréal H4H 1R3, Canada
- Department of Psychiatry, McGill University, Montréal H3A 1A1, Canada
| | - Maria Natasha Rajah
- Douglas Mental Health University Institute Research Centre, McGill University, Montréal H4H 1R3, Canada
- Department of Psychiatry, McGill University, Montréal H3A 1A1, Canada
- Department of Psychology, Toronto Metropolitan University, Toronto M5S 1A1, Canada
| | - Sylvia Villeneuve
- Douglas Mental Health University Institute Research Centre, McGill University, Montréal H4H 1R3, Canada
- Department of Psychiatry, McGill University, Montréal H3A 1A1, Canada
| | - Anne Maass
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg 39120, Germany
- Institute for Biology, Otto von Guericke University Magdeburg, Magdeburg 39120, Germany
| |
Collapse
|