1
|
Qi Z, Li Z, Shan P, Wang Q, Sun W. SSA-classifier based screening study for Alzheimer's disease. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 339:126115. [PMID: 40273763 DOI: 10.1016/j.saa.2025.126115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 03/18/2025] [Accepted: 03/25/2025] [Indexed: 04/26/2025]
Abstract
Alzheimer's is a disease (AD) that affects 10 % of individuals aged ≥ 65, is the most prevalent neurodegenerative disorder. We propose a diagnostic framework integrating plasma attenuated total reflection Fourier transform infrared (ATR-FTIR) spectroscopy with machine learning for AD screening. Four classifiers (SVM, Logistic Regression, XGBoost, LDA) were optimized using a modified Sparrow Search Algorithm (GSSA), benchmarked against its standard version (SSA) and Bayesian methods. GSSA-optimized classifiers demonstrated superior performance, with GSSA-XGBoost achieving peak metrics: 88.51 % accuracy (+2.30 % vs SSA-XGBoost), 95.35 % sensitivity, and 81.82 % specificity. Comparative test-set results revealed consistent improvements: SSA-optimized models attained 83.91 % (SVM), 77.01 % (Logistic), 86.21 % (XGBoost), and 79.31 % (LDA) accuracy, and Bayesian counterparts achieved 85.06 %, 80.46 %, 85.06 %, and 79.31 %,while GSSA-optimized models achieved 86.21 %,80.46 %,88.51 %,80.46 %,respectively. Moreover, GSSA further enhanced sensitivities to 97.67 % (SVM/LDA) and specificities to 81.82 % (XGBoost), outperforming both SSA and Bayesian approaches. This ATR-FTIR/GSSA-machine learning synergy shows significant potential as a minimally invasive AD screening tool, with XGBoost delivering optimal diagnostic balance. Our methodology advances spectroscopic biomarker discovery while demonstrating algorithmic optimization efficacy.
Collapse
Affiliation(s)
- Zihao Qi
- School of Control Engineering, Northeastern University at Qinhuangdao, Qinhuangdao 066003, China
| | - Zhigang Li
- Northeastern University at Qinhuangdao, Qinhuangdao 066003, China.
| | - Peng Shan
- Northeastern University at Qinhuangdao, Qinhuangdao 066003, China
| | - Qiaoyun Wang
- Northeastern University at Qinhuangdao, Qinhuangdao 066003, China
| | - Weishang Sun
- School of Electrical Engineering, Yanshan University, Qinhuangdao 066003, China
| |
Collapse
|
2
|
Su C, Huang T, Zhang M, Zhang Y, Zeng Y, Chen X. Glucocorticoid receptor signaling in the brain and its involvement in cognitive function. Neural Regen Res 2025; 20:2520-2537. [PMID: 39248182 PMCID: PMC11801288 DOI: 10.4103/nrr.nrr-d-24-00355] [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: 04/01/2024] [Revised: 06/07/2024] [Accepted: 07/06/2024] [Indexed: 09/10/2024] Open
Abstract
The hypothalamic-pituitary-adrenal axis regulates the secretion of glucocorticoids in response to environmental challenges. In the brain, a nuclear receptor transcription factor, the glucocorticoid receptor, is an important component of the hypothalamic-pituitary-adrenal axis's negative feedback loop and plays a key role in regulating cognitive equilibrium and neuroplasticity. The glucocorticoid receptor influences cognitive processes, including glutamate neurotransmission, calcium signaling, and the activation of brain-derived neurotrophic factor-mediated pathways, through a combination of genomic and non-genomic mechanisms. Protein interactions within the central nervous system can alter the expression and activity of the glucocorticoid receptor, thereby affecting the hypothalamic-pituitary-adrenal axis and stress-related cognitive functions. An appropriate level of glucocorticoid receptor expression can improve cognitive function, while excessive glucocorticoid receptors or long-term exposure to glucocorticoids may lead to cognitive impairment. Patients with cognitive impairment-associated diseases, such as Alzheimer's disease, aging, depression, Parkinson's disease, Huntington's disease, stroke, and addiction, often present with dysregulation of the hypothalamic-pituitary-adrenal axis and glucocorticoid receptor expression. This review provides a comprehensive overview of the functions of the glucocorticoid receptor in the hypothalamic-pituitary-adrenal axis and cognitive activities. It emphasizes that appropriate glucocorticoid receptor signaling facilitates learning and memory, while its dysregulation can lead to cognitive impairment. This provides clues about how glucocorticoid receptor signaling can be targeted to overcome cognitive disability-related disorders.
Collapse
Affiliation(s)
- Chonglin Su
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Taiqi Huang
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Meiyu Zhang
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Yanyu Zhang
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Yan Zeng
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| | - Xingxing Chen
- Brain Science and Advanced Technology Institute, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
3
|
Povedano E, Garranzo-Asensio M, Montero-Calle A, Valverde A, Dalmasso P, San Segundo-Acosta P, Cano O, Vázquez M, Mas V, Fernández-Aceñero MJ, Rivas G, Pingarrón JM, Campuzano S, Barderas R. Novel 6xHis/HaloTag mammalian expressed autoantigens for the detection of humoral response with multiplexed electrochemical biosensors: A breakthrough in colorectal cancer and Alzheimer's disease personalized diagnostics. Biosens Bioelectron 2025; 282:117506. [PMID: 40288309 DOI: 10.1016/j.bios.2025.117506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 04/02/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
The integration of autoantibody detection with electrochemical biosensors allows the development of a novel strategy for managing colorectal cancer (CRC) and Alzheimer's disease (AD). This work reports the use, for the first time, of sustainable receptors, autoantigens expressed in mammalian cells, fused to 6xHis at the N-terminus and HaloTag at the C-terminus, immobilized on chloroalkane-modified magnetic beads (MBs) to selectively capture plasma autoantibodies. Detection was based on amperometric measurements using disposable multiplexed screen-printed carbon electrodes (x8 sensing surfaces), horseradish peroxidase (HRP)-conjugated secondary antibodies, and the HQ/H2O2 system. HaloTag immobilization gave rise to a great sensitivity allowing discrimination between patients and healthy individuals in comparison with HisTag or -COOH immobilization. The CRC biosensor integrated seven CRC-specific full-length autoantigens, while the AD bioplatform combined three AD-specific full-length proteins and four AD-specific peptides, which allowed a robust diagnostic performance validated by ROC analyses. These multiplexed biosensors provide minimally invasive, cost-effective, and sustainable alternatives to traditional diagnostic methods, and show ideal for discovery of new targets and for mass screening and early disease detection, supporting personalized medicine approaches.
Collapse
Affiliation(s)
- Eloy Povedano
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, Plaza de Las Ciencias 2, 28040, Madrid, Spain
| | - María Garranzo-Asensio
- Chronic Disease Programme, UFIEC, Institute of Health Carlos III, Majadahonda, 28220, Madrid, Spain
| | - Ana Montero-Calle
- Chronic Disease Programme, UFIEC, Institute of Health Carlos III, Majadahonda, 28220, Madrid, Spain
| | - Alejandro Valverde
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, Plaza de Las Ciencias 2, 28040, Madrid, Spain
| | - Pablo Dalmasso
- Research and Transfer Center in Environmental Chemical Engineering, CONICET, Córdoba Regional Faculty, National Technological University, Maestro López Esq. Cruz Roja Argentina, 5016, Córdoba, Argentina
| | - Pablo San Segundo-Acosta
- Chronic Disease Programme, UFIEC, Institute of Health Carlos III, Majadahonda, 28220, Madrid, Spain
| | - Olga Cano
- Respiratory Viruses Laboratory, National Center for Microbiology Instituto de Salud Carlos III Majadahonda, 28220, Madrid, Spain
| | - Mónica Vázquez
- Respiratory Viruses Laboratory, National Center for Microbiology Instituto de Salud Carlos III Majadahonda, 28220, Madrid, Spain
| | - Vicente Mas
- Respiratory Viruses Laboratory, National Center for Microbiology Instituto de Salud Carlos III Majadahonda, 28220, Madrid, Spain
| | - María Jesús Fernández-Aceñero
- Department of Legal Medicine, Psychiatry and Surgical Pathology, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Clínico San Carlos (IdISCC), Surgical Pathology Department, Hospital Universitario Clínico San Carlos, E-28040, Madrid, Spain
| | - Gustavo Rivas
- Institute of Physical-Chemical Research of Córdoba, CONICET-UNC, Faculty of Chemical Sciences, National University of Córdoba, University City, 5000, Córdoba, Argentina
| | - José M Pingarrón
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, Plaza de Las Ciencias 2, 28040, Madrid, Spain.
| | - Susana Campuzano
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, Plaza de Las Ciencias 2, 28040, Madrid, Spain; CIBER of Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, 28046, Madrid, Spain.
| | - Rodrigo Barderas
- Chronic Disease Programme, UFIEC, Institute of Health Carlos III, Majadahonda, 28220, Madrid, Spain; CIBER of Frailty and Healthy Aging (CIBERFES), Instituto de Salud Carlos III, 28046, Madrid, Spain.
| |
Collapse
|
4
|
Wei R, Buss SS, Milde R, Fernandes M, Sumsion D, Davis E, Kong WY, Xiong Y, Veltink J, Rao S, Westover TM, Petersen L, Turley N, Singh A, Das S, Junior VM, Ghanta M, Gupta A, Kim J, Lam AD, Stone KL, Mignot E, Hwang D, Trotti LM, Clifford GD, Katwa U, Thomas RJ, Mukerji S, Zafar SF, Westover MB, Sun H. Automated phenotyping of mild cognitive impairment and Alzheimer's disease and related dementias using electronic health records. Int J Med Inform 2025; 200:105917. [PMID: 40222334 DOI: 10.1016/j.ijmedinf.2025.105917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 01/31/2025] [Accepted: 04/07/2025] [Indexed: 04/15/2025]
Abstract
OBJECTIVES Unstructured and structured data in electronic health records (EHR) are a rich source of information for research and quality improvement studies. However, extracting accurate information from EHR is labor-intensive. Timely and accurate identification of patients with Alzheimer's Disease, related dementias (ADRD), or mild cognitive impairment (MCI) is critical for improving patient outcomes through early intervention, optimizing care plans, and reducing healthcare system burdens. Here we introduce an automated EHR phenotyping model to streamline this process and enable efficient identification of these conditions. METHODS We analyzed data from 3,626 outpatients seen at two hospitals between February 2015 and June 2022. Through manual chart review, we established ground truth labels for the presence or absence of MCI/ADRD diagnoses. Our model combined three types of data: (1) unstructured clinical notes, from which we extracted single words, two-word phrases (bigrams), and three-word phrases (trigrams) as features, weighted using Term Frequency-Inverse Document Frequency (TF-IDF) to capture their relative importance, (2) International Classification of Diseases (ICD) codes, and (3) medication prescriptions related to MCI/ADRD. We trained a regularized logistic regression model to predict MCI/ADRD diagnoses and evaluated its performance using standard metrics including area under the receiver operating curve (AUROC), area under the precision-recall curve (AUPRC), accuracy, specificity, precision, recall, and F1 score. RESULTS Thirty percent of patients in the cohort carried diagnoses of MCI/ADRD based on manual review. When evaluated on a held-out test set, the best model using clinical notes, ICDs, and medications, achieved an AUROC of 0.98, an AUPRC of 0.98, an accuracy of 0.93, a sensitivity (recall) of 0.91, a specificity of 0.96, a precision of 0.96, and an F1 score of 0.93 The estimated overall accuracy for patients randomly selected from EHRs was 99.88%. CONCLUSION Automated EHR phenotyping accurately identifies patients with MCI/ADRD based on clinical notes, ICD codes, and medication records. This approach holds potential for large-scale MCI/ADRD research utilizing EHR databases.
Collapse
Affiliation(s)
- Ruoqi Wei
- Beth Israel Deaconess Medical Center, Boston, MA, USA; Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, FL, USA
| | - Stephanie S Buss
- Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Rebecca Milde
- Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Marta Fernandes
- Beth Israel Deaconess Medical Center, Boston, MA, USA; Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Daniel Sumsion
- Beth Israel Deaconess Medical Center, Boston, MA, USA; Boston Children's Hospital, Boston, MA, USA
| | - Elijah Davis
- Beth Israel Deaconess Medical Center, Boston, MA, USA; Brigham Young University, Provo, UT 84602, USA
| | - Wan-Yee Kong
- Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Yiwen Xiong
- Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Jet Veltink
- Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Samvrit Rao
- Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Tara M Westover
- Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Lydia Petersen
- Beth Israel Deaconess Medical Center, Boston, MA, USA; Boston Children's Hospital, Boston, MA, USA
| | - Niels Turley
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Arjun Singh
- Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Sudeshna Das
- Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Valdery Moura Junior
- Beth Israel Deaconess Medical Center, Boston, MA, USA; Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Manohar Ghanta
- Beth Israel Deaconess Medical Center, Boston, MA, USA; Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Aditya Gupta
- Beth Israel Deaconess Medical Center, Boston, MA, USA; Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | | | - Alice D Lam
- Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Katie L Stone
- California Pacific Medical Center Research Institute, San Francisco, CA, USA; Department of Epidemiology and Biostatistics, University of California, San Francisco, USA
| | | | - Dennis Hwang
- Kaiser Permanente, San Bernardino County Sleep Disorders Center. San Bernardino, CA, USA
| | | | - Gari D Clifford
- Emory University School of Medicine, Atlanta, GA, USA; Georgia Institute of Technology, Atlanta, GA, USA
| | - Umakanth Katwa
- Harvard Medical School, Boston, MA, USA; Boston Children's Hospital, Boston, MA, USA
| | - Robert J Thomas
- Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Shibani Mukerji
- Beth Israel Deaconess Medical Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Sahar F Zafar
- Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - M Brandon Westover
- Beth Israel Deaconess Medical Center, Boston, MA, USA; Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Haoqi Sun
- Beth Israel Deaconess Medical Center, Boston, MA, USA; Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Stojanovic M, Waters AB, Kiselica AM, Benge JF. The impact of technology-based compensatory behaviors on subjective cognitive decline in older adults with a family history of dementia. APPLIED NEUROPSYCHOLOGY. ADULT 2025; 32:1162-1170. [PMID: 37647340 DOI: 10.1080/23279095.2023.2247109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
The current study examined whether greater use of technology to help with daily tasks is associated with less subjective cognitive decline (SCD), especially in individuals with a family history of Alzheimer's disease (AD). Individuals over the age of 50 (n = 102; age range 50-85) completed surveys about their digital and analog approaches to daily tasks, physical activity, and SCD. Participants with and without family histories of AD were matched on age, education, sex, and family history of AD using the R package MatchIt. There was no main effect of technology-based behavioral strategies on SCD (p = 0.259). However, a family history of AD moderated the association between technology use and SCD even when controlling for another protective lifestyle factor, physical activity. In individuals with a family history of AD, more reliance on technology-based behavioral strategies was associated with less SCD (p = 0.018), but this relationship was not significant in individuals without family history of AD (p = 0.511). Our findings suggest that technology-based behavioral strategies are associated with less SCD in individuals with a family history of AD, independent of another protective lifestyle factor. Future recommendations provided by healthcare providers to address SCD in cognitively unimpaired older adults might include focusing on technological assistance.
Collapse
Affiliation(s)
- Marta Stojanovic
- Department of Health Psychology, University of Missouri, Columbia, MO, USA
- Department of Psychological and Brain Sciences, Washington University in St. Louis, Saint Louis, MO, USA
| | - Abigail B Waters
- Department of Health Psychology, University of Missouri, Columbia, MO, USA
- Department of Psychology, Suffolk University, Boston, MA, USA
| | - Andrew M Kiselica
- Department of Health Psychology, University of Missouri, Columbia, MO, USA
| | - Jared F Benge
- Department of Neurology, University of Texas at Austin, Austin, TX, USA
- Mulva Clinic for the Neurosciences, University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
6
|
Lemche E, Hortobágyi T, Kiecker C, Turkheimer F. Neuropathological links between T2DM and LOAD: systematic review and meta-analysis. Physiol Rev 2025; 105:1429-1486. [PMID: 40062731 DOI: 10.1152/physrev.00040.2024] [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: 10/01/2024] [Revised: 02/01/2025] [Accepted: 02/22/2025] [Indexed: 04/16/2025] Open
Abstract
Recent decades have described parallel neuropathological mechanisms increasing the risk for developing late-onset Alzheimer's dementia (LOAD) in type 2 diabetes mellitus (T2DM); however, still little is known of the role of diabetic encephalopathy and brain atrophy in LOAD. The aim of this systematic review is to provide a comprehensive view on diabetic encephalopathy/cerebral atrophy, taking into account neuroimaging data, neuropathology, metabolic and endocrine mechanisms, amyloid formation, brain perfusion impairments, neuroimmunology, and inflammasome activation. Key switches were identified, to further meta-analyze genomic candidate loci and epigenetic modifications. For the qualitative meta-analysis of genomic bases extracted, human linkage studies were examined; for epigenetic mechanisms, data from both human and animal studies are described. For the systematic review of pathophysiological mechanisms, 1,259 publications were evaluated and 93 gene loci extracted for candidate risk linkages. Sixty-six publications were evaluated for genomic association and descriptions of epigenomic modifications. Overall accumulated results highlight the insulin signaling system, vascular markers, inflammation and inflammasome pathways, amylin interactions, and glycosylation mechanisms. The protocol was registered with PROSPERO (ID: CRD42023440535).
Collapse
Affiliation(s)
- Erwin Lemche
- Section of Cognitive Neuropsychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Tibor Hortobágyi
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
- Department of Neurology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Clemens Kiecker
- Department for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Federico Turkheimer
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
7
|
Piccolino AL, Piccolino AR, Piccolino SG. Distinguishing Alzheimer's disease from other dementias using pattern profile analysis in the Meyers Neuropsychological Battery: An exploratory study. APPLIED NEUROPSYCHOLOGY. ADULT 2025; 32:1087-1102. [PMID: 37477644 DOI: 10.1080/23279095.2023.2236742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Abstract
OBJECTIVE This exploratory study aimed to assess the efficacy of pattern-matching statistical methods within the Meyers Neuropsychological Battery (MNB). It compared neuropsychological test data profiles of Alzheimer's disease (AD) patients from three independent samples against four MNB dementia groups: MNB-AD, MNB-Vascular Dementia (VaD), MNB-Dementia with Lewy bodies (DLB), and MNB-Parkinson's disease dementia (PDD). MATERIALS AND METHODS Three AD-independent samples completed either the MNB (referred to as I-MNB-AD), Dementia Rating Scale-2 with additional testing (denoted as DRS-Plus-AD), or the Repeatable Battery for the Assessment of Neuropsychological Status (designated as RBANS-AD). Test data profiles were cross-validated with four MNB dementia comparison group datasets. Statistical methods included Pearson correlation, Kullback-Leibler (KL) divergence, pooled effect size (Cohen's d), Configuration, and MNB Code. RESULTS Classification accuracy ranged from 40% (Pearson r) to 88% (Cohen's d) in the I-MNB-AD sample, 47% (Cohen's d) to 93% (KL) in the DRS-Plus-AD sample, and 47% (Pearson r) to 78% (Configuration) in the RBANS-AD sample. Some methods showed limited effectiveness depending on the sample and comparison group analyzed, while others demonstrated strong performance. Using a simple majority count of agreement, classification rates for selecting the MNB-AD comparison group were 80% (I-MNB-AD), 85% (DRS-Plus-AD), and 66% (RBANS-AD). CONCLUSIONS This exploratory study demonstrates that specific statistical methods employed in the MNB for pattern-matching analysis effectively differentiated neuropsychological profiles of individuals with AD from other types of dementia, contributing to improved diagnostic precision. The findings underscore the potential advantages of pattern-matching analysis, advocating for further research to validate and refine its application.
Collapse
|
8
|
Lorentzen IM, Espenes J, Eliassen IV, Hessen E, Waterloo K, Nakling A, Gísladóttir B, Jarholm J, Fladby T, Kirsebom BE. Investigating the relationship between allocentric spatial working memory and biomarker status in preclinical and prodromal Alzheimer's disease. APPLIED NEUROPSYCHOLOGY. ADULT 2025; 32:1074-1086. [PMID: 37552673 DOI: 10.1080/23279095.2023.2236262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/10/2023]
Abstract
The 4 Mountain Test (4MT) is a test of allocentric spatial working memory and has been proposed as an earlier marker of predementia Alzheimer's disease (AD) than episodic verbal memory. We here compare the 4MT to the CERAD word list memory recall in both cognitively normal (CN) and mild cognitive impairment (MCI) cases with or without cerebrospinal fluid markers (CSF) of Alzheimer's disease pathology. Linear regression was used to assess the influence of CSF determined Aβ-plaque (Aβ-/+) or neurofibrillary tau tangles (Tau-/+) on 4MT and CERAD recall performance. Analyses were performed in the full sample and the CN and MCI sub-samples. Pearson correlations were calculated to examine the relationship between 4MT and tests of psychomotor speed, verbal memory, cognitive flexibility, verbal fluency, and visuo-spatial perception. Analyses showed no significant differences in 4MT scores between Aβ-/Aβ+, nor Tau-/Tau + participants, irrespective of cognitive status. In contrast, CERAD recall scores were lower in both Aβ+ compared to Aβ- (p<.01), and Tau + compared to Tau- participants (p<.01) in the full sample analyses. There were no significant differences in CERAD recall performance between Aβ- vs. Aβ+ and Tau- vs. to Tau + in the in CN/MCI sub-samples. 4MT scores were significantly correlated with tests of psychomotor speed, cognitive flexibility, and visuo-spatial perception in the full sample analyses. In conclusion, the CERAD recall outperformed the 4MT as a cognitive marker of CSF determined AD pathology. This suggests that allocentric working memory, as measured by the 4MT, may not be used as an early marker of predementia AD.
Collapse
Affiliation(s)
- Ingrid Myrvoll Lorentzen
- Department of Psychology, Faculty of Health Sciences, The Arctic University of Norway, Tromsø, Norway
| | - Jacob Espenes
- Department of Psychology, Faculty of Health Sciences, The Arctic University of Norway, Tromsø, Norway
| | - Ingvild Vøllo Eliassen
- Department of Psychology, University of Oslo, Oslo, Norway
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway
| | - Erik Hessen
- Department of Psychology, University of Oslo, Oslo, Norway
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway
| | - Knut Waterloo
- Department of Psychology, Faculty of Health Sciences, The Arctic University of Norway, Tromsø, Norway
- Department of Neurology, University Hospital of North Norway, Tromsø, Norway
| | - Arne Nakling
- Institute of Clinical Medicine, University of Bergen, Norway
| | - Berglind Gísladóttir
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway
- Department of Clinical Molecular Biology (EpiGen), University of Oslo and Akershus University Hospital, Lørenskog, Norway
| | - Jonas Jarholm
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Tormod Fladby
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Bjørn-Eivind Kirsebom
- Department of Psychology, Faculty of Health Sciences, The Arctic University of Norway, Tromsø, Norway
- Department of Neurology, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|
9
|
Bonarota S, Caruso G, Domenico CD, Sperati S, Tamigi FM, Giulietti G, Giove F, Caltagirone C, Serra L. Integration of automatic MRI segmentation techniques with neuropsychological assessments for early diagnosis and prognosis of Alzheimer's disease. A systematic review. Neuroimage 2025; 314:121264. [PMID: 40368056 DOI: 10.1016/j.neuroimage.2025.121264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 04/24/2025] [Accepted: 05/08/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND This systematic review investigates the integration of automatic segmentation techniques of magnetic resonance imaging (MRI) with neuropsychological assessments for early diagnosis and prognosis of Alzheimer's Disease (AD). OBJECTIVES Focus on studies that utilise automated MRI segmentation and neuropsychological evaluations across the AD spectrum. DATA SOURCES A literature search was conducted on the PubMed database on 7 November 2024, using key terms related to MRI, segmentation, brain structures, AD, and cognitive decline. STUDY ELIGIBILITY CRITERIA Studies including individuals with AD, mild cognitive impairment (MCI), or subjective cognitive decline (SCD), utilising structural MRI, focusing on grey matter and automatic segmentation, and reporting cognitive assessments were included. STUDY APPRAISAL AND SYNTHESIS METHODS Data were extracted and synthesised focusing on associations between MRI measures and cognitive tests, and discriminative values for diagnosis or prognosis. RESULTS Seven studies were included, showing a significant association between structural changes in the medial temporal lobe and cognitive decline. The combination of MRI volumetric measures and neuropsychological scores enhanced diagnostic accuracy. Neuropsychological measures demonstrated superiority in the identification of patients with MCI and mild AD in comparison to MRI measures alone. LIMITATIONS Heterogeneity across studies, selection and measurement bias, and lack of non-response data were noted. CONCLUSIONS AND IMPLICATIONS This review emphasises the necessity of integrating automated MRI segmentation with neuropsychological assessments for the diagnosis and prognosis of AD. While MRI is valuable, neuropsychological testing remains essential for early detection. Future studies should focus on developing integrated predictive models and refining neuroimaging techniques.
Collapse
Affiliation(s)
- Sabrina Bonarota
- Neuroimaging Laboratory, Fondazione Santa Lucia IRCCS, Rome, Italy
| | - Giulia Caruso
- SC Neurologia Ospedaliera, Policlinico Riuniti di Foggia, Foggia, Italy
| | - Carlotta Di Domenico
- Neuroimaging Laboratory, Fondazione Santa Lucia IRCCS, Rome, Italy; Department of Psychology, Sapienza University of Rome
| | - Sofia Sperati
- Neuroimaging Laboratory, Fondazione Santa Lucia IRCCS, Rome, Italy; Department of Psychology, Sapienza University of Rome
| | - Federico Maria Tamigi
- Neuroimaging Laboratory, Fondazione Santa Lucia IRCCS, Rome, Italy; Department of Psychology, Sapienza University of Rome
| | - Giovanni Giulietti
- Neuroimaging Laboratory, Fondazione Santa Lucia IRCCS, Rome, Italy; Department of Human Anatomy-Histology-Forensic Medicine-Orthopedics Sapienza University of Rome
| | - Federico Giove
- Neuroimaging Laboratory, Fondazione Santa Lucia IRCCS, Rome, Italy; Museo Storico della Fisica e Centro Studi e Ricerche Enrico Fermi, Rome, Italy
| | | | - Laura Serra
- Neuroimaging Laboratory, Fondazione Santa Lucia IRCCS, Rome, Italy; Department of Human Sciences, Guglielmo Marconi University, Rome, Italy.
| |
Collapse
|
10
|
Barros-Aragão FG, Pinheiro TL, Pinto TP, Vanderborgh B, Rezende NB, de Freitas GB, de Freitas GR, Bozza FA, Souza AS, Rodrigues EC, Brandão CO, Mattos P, Sudo FK, Tovar-Moll FF, De Felice FG. Comparison of cerebrospinal fluid biomarkers in patients with severe COVID-19 neurological outcomes and Alzheimer's disease. Brain Behav Immun Health 2025; 46:101007. [PMID: 40417395 PMCID: PMC12099766 DOI: 10.1016/j.bbih.2025.101007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 03/26/2025] [Accepted: 04/29/2025] [Indexed: 05/27/2025] Open
Abstract
Background COVID-19 induces acute and long-term neurological symptoms. Links between COVID-19 neurological disturbance and Alzheimer's disease (AD) have been hypothesized because neuroinflammation plays a significant role in both diseases. However, it is unknown if COVID-19 patients with neurological disturbance present molecular alterations related to AD pathology. A better understanding of possible molecular links between COVID-19-induced neurological disease and AD would lead to improved patient follow-up and late-onset disease prevention. Here, we analyze early AD biomarkers in a Brazilian cohort of COVID-19 patients with neurological symptoms. We compared COVID-19 patients' neuroinflammatory and AD biomarker levels to controls, amnestic mild cognitive impairment (aMCI), and AD. Methods We analyzed cerebrospinal (CSF) biomarkers of neuroinflammation (interleukin-6 (IL6)), amyloid-beta (Aβ) proteinopathy (Aβ42/40), phosphorylated Tau (pTau181), and the neurodegeneration-associated biomarker total Tau in controls (n = 36), COVID-19 patients presenting neurological alterations (n = 35), aMCI (n = 19), and AD patients (n = 20). Comparisons were corrected by possible sex, age, and comorbidities confounding effects. CSF biomarkers were correlated with systemic and neuro-inflammation markers. Results We found that severe COVID-19 patients presented higher CSF Tau than controls, comparable to alterations observed in AD patients. However, we did not find changes in CSF Aβ42/40, pTau-181/Aβ42, or Tau/Aβ42 ratios. Severe COVID-19 patients presented higher Tau, Tau/Aβ42, and pTau181/Aβ42 than mild patients. In COVID-19 patients, CSF pro-inflammatory cytokine IL6 and AD biomarkers correlated with systemic inflammatory index (SII). Conclusions Collectively, our findings reveal that CSF tau levels are comparably elevated in COVID-19 neurological patients and AD, suggesting ongoing neurodegeneration in COVID-19 neurological disease, but no biomarker alterations related to AD pathology. Furthermore, CNS AD-related biomarker levels in COVID-19 patients change in association with disease severity and systemic inflammation. Considering that inflammation may persist post-COVID, our findings urge the assessment of possible AD-related biomarker changes in COVID-19 survivors with lingering symptoms.
Collapse
Affiliation(s)
- Fernanda G.Q. Barros-Aragão
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences & Department of Psychiatry, Queen’s University, Kingston, Ontario, Canada, K7L 3N6
- Institute of Medical Biochemistry Leopoldo De Meis (IBqM), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil, 21941-902
| | - Thaís L. Pinheiro
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
- Institute of Medical Biochemistry Leopoldo De Meis (IBqM), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil, 21941-902
| | - Talita P. Pinto
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
| | - Bart Vanderborgh
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
| | - Nathane B.S. Rezende
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
| | - Guilherme B. de Freitas
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences & Department of Psychiatry, Queen’s University, Kingston, Ontario, Canada, K7L 3N6
- Institute of Medical Biochemistry Leopoldo De Meis (IBqM), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil, 21941-902
| | | | - Fernando A. Bozza
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
| | - Andrea S. Souza
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
| | - Erika C. Rodrigues
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
| | | | - Paulo Mattos
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
| | - Felipe K. Sudo
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
| | | | - Fernanda G. De Felice
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences & Department of Psychiatry, Queen’s University, Kingston, Ontario, Canada, K7L 3N6
- Institute of Medical Biochemistry Leopoldo De Meis (IBqM), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil, 21941-902
| |
Collapse
|
11
|
Shrestha S, Zhu X, Griswold ME, Palta P, Sullivan KJ, Chen H, Schneider ALC, Moghekar A, Grove ML, Thyagarajan B, Pike JR, Gottesman RF, Windham BG, Mosley TH, Deal JA, Kamath V. Olfaction and Plasma Biomarkers of Alzheimer Disease and Neurodegeneration in the Atherosclerosis Risk in Communities Study. Neurology 2025; 104:e213706. [PMID: 40373252 DOI: 10.1212/wnl.0000000000213706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/26/2025] [Indexed: 05/17/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Investigation of olfactory impairment, an early manifestation of Alzheimer disease (AD), in relation to plasma biomarkers of AD and neurodegeneration could provide insights into the disease's pathophysiology. Because few such studies based on large, diverse, community-based populations exist, we investigated associations of odor identification ability with plasma biomarkers of AD and other neurodegenerative pathologies in community-dwelling Black and White older adults. METHODS This cross-sectional investigation included participants from the Atherosclerosis Risk in Communities study who attended visit 5 (2011-2013) and who completed olfactory testing and brain MRI examinations and had plasma biomarkers measured (namely, amyloid-beta [Aβ]42/Aβ40 ratio, phosphorylated-tau at threonine-181 [p-tau181], p-tau181/Aβ42 ratio, glial fibrillary acidic protein [GFAP], and neurofilament light chain [NfL]). Odor identification ability was measured by the 12-item Sniffin' Sticks test. Separate linear regression models were used to examine the association of continuous olfaction score and olfaction categories (anosmia: score 6; hyposmia: 7-8; moderate-normal: 9-10; good-normal: 11-12) with each biomarker (all were log-transformed), adjusting for sociodemographic and cardiovascular factors, head injury, APOE-ε4 status, and estimated glomerular filtration rate. We further examined whether any observed associations are explained by total and regional brain volumes. RESULTS Among 1,545 participants (age: 76 ± 5 years, 60% women, 27% self-reported Black participants), the mean olfaction score was 9.2 ± 2.3; 14% had anosmia. Consistent with our hypotheses, poorer olfactory scores were associated with higher plasma p-tau181 (β per 1-unit lower score: 0.026 [95% CI 0.012-0.040]), p-tau181/Aβ42 (β: 0.027 [95% CI 0.011- 0.044]), GFAP (β: 0.024 [95% CI 0.009-0.040]), and NfL (β: 0.034 [95% CI 0.019-0.050] and lower Aβ42/Aβ40 ratio (β: -0.007 [95% CI -0.015 to 0.000]). Likewise, compared with good-normal olfaction, anosmia showed associations with all biomarker levels indicative of greater neuropathology (e.g., β for plasma p-tau181/Aβ42: 0.235 [95% CI 0.113-0.358] and β for plasma NfL: 0.210 [95% CI 0.102-0.317]), although the association with Aβ42/Aβ40 ratio was not statistically significant (β: -0.054 [95% CI -0.108 to 0.001]). These biomarkers were not significantly associated with hyposmia or moderate-normal olfaction. Smaller medial-temporal lobe volumes partly explained olfaction's link with plasma p-tau181, p-tau181/Aβ42, GFAP, and NfL. DISCUSSION Our findings suggest that poor olfaction is associated with multiple AD-related and other neurodegenerative processes. Future studies should investigate how longitudinal changes in both olfaction and biomarkers relate to each other.
Collapse
Affiliation(s)
- Srishti Shrestha
- The Memory Impairment and Neurodegenerative Dementia (MIND) Center, University of Mississippi Medical Center, Jackson, MS
| | - Xiaoqian Zhu
- The Memory Impairment and Neurodegenerative Dementia (MIND) Center, University of Mississippi Medical Center, Jackson, MS
| | - Michael E Griswold
- The Memory Impairment and Neurodegenerative Dementia (MIND) Center, University of Mississippi Medical Center, Jackson, MS
| | - Priya Palta
- Department of Neurology, University of North Carolina at Chapel Hill, NC
| | - Kevin J Sullivan
- The Memory Impairment and Neurodegenerative Dementia (MIND) Center, University of Mississippi Medical Center, Jackson, MS
| | - Honglei Chen
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI
| | - Andrea Lauren Christman Schneider
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Megan L Grove
- Human Genetics Center, Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, TX
| | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis
| | - James Russell Pike
- Optimal Aging Institute, New York University Grossman School of Medicine, NY
| | - Rebecca F Gottesman
- Stroke Branch, National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, MD
| | - B Gwen Windham
- The Memory Impairment and Neurodegenerative Dementia (MIND) Center, University of Mississippi Medical Center, Jackson, MS
| | - Thomas H Mosley
- The Memory Impairment and Neurodegenerative Dementia (MIND) Center, University of Mississippi Medical Center, Jackson, MS
| | - Jennifer A Deal
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, MD; and
| | - Vidyulata Kamath
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
12
|
Sbriscia M, Spadoni T, Ambrogini P, Guescini M, Agostini R, Graciotti L, Piacenza F, Giuli C, Cecati M, Bonfigli AR, Vaiasicca S, Pagliarini M, Rusanova I, Fazioli F, Sabbatinelli J, Albertini MC, Olivieri F, Giuliani A. miR-132-3p is down-regulated in plasma and CD171 + extracellular vesicles isolated from patients with mild Alzheimer's disease. Mech Ageing Dev 2025; 225:112063. [PMID: 40252983 DOI: 10.1016/j.mad.2025.112063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/28/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025]
Abstract
Alzheimer's disease (AD), the most prevalent neurodegenerative disorder in aging populations, demands minimally invasive biomarkers for early diagnosis and monitoring. Circulating microRNAs (miRNAs) show promise as such biomarkers. In this study, we examined the levels of five selected miRNAs, implicated in neurodegenerative processes, in plasma and neuron-derived extracellular vesicles (EVs) from cognitively healthy controls (n = 5), and patients with mild (n = 10) and moderate AD (n = 10), stratified by Mini-Mental State Examination (MMSE). miR-23a-3p, miR-223a-3p, and miR-132-3p were significantly downregulated in both plasma and EVs of AD patients, with miR-132-3p emerging as the strongest biomarker candidate for mild AD. Plasma miRNA levels strongly correlated with EV cargo, supporting plasma-based assessments. To validate these findings, miR-132-3p levels were analyzed in expanded cohorts, including cognitively healthy subjects (n = 36), mild AD (n = 37), and moderate AD (n = 40), as well as a cohort of subjects with mild cognitive impairment (MCI, n = 31) and an additional external cohort of cognitively healthy subjects (CTR external, n = 37). Results confirmed miR-132-3p downregulation in AD patients and revealed a significant elevation in MCI individuals, suggesting a potential neuroprotective role in AD early stages. These findings highlight miR-132-3p as a promising, minimally invasive biomarker for early AD diagnosis and disease progression monitoring.
Collapse
Affiliation(s)
- Matilde Sbriscia
- Clinic of Laboratory and Precision Medicine, IRCCS INRCA, Ancona, Italy
| | - Tatiana Spadoni
- Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, Ancona, Italy
| | - Patrizia Ambrogini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Michele Guescini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Rachele Agostini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Laura Graciotti
- Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, Ancona, Italy
| | - Francesco Piacenza
- Advanced Technology Center for Aging Research, IRCCS INRCA, Ancona, Italy
| | - Cinzia Giuli
- Geriatric Operative Unit, IRCCS INRCA, Fermo, Italy
| | - Monia Cecati
- Advanced Technology Center for Aging Research, IRCCS INRCA, Ancona, Italy
| | | | | | | | - Iryna Rusanova
- Departamento de Bioquímica y Biología Molecular I, Facultad de Ciencias, Instituto de Biotecnología, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Granada, Spain
| | - Francesca Fazioli
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Ancona, Italy
| | - Jacopo Sabbatinelli
- Clinic of Laboratory and Precision Medicine, IRCCS INRCA, Ancona, Italy; Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Ancona, Italy.
| | | | - Fabiola Olivieri
- Clinic of Laboratory and Precision Medicine, IRCCS INRCA, Ancona, Italy; Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Ancona, Italy
| | - Angelica Giuliani
- Cardiac Rehabilitation Unit of Bari Institute, Istituti Clinici Scientifici Maugeri IRCCS, Bari, Italy
| |
Collapse
|
13
|
Jiang J, Zhao K, Li W, Zheng P, Jiang S, Ren Q, Duan Y, Yu H, Kang X, Li J, Hu K, Jiang T, Zhao M, Wang L, Yang S, Zhang H, Liu Y, Wang A, Liu Y, Xu J. Multiomics Reveals Biological Mechanisms Linking Macroscale Structural Covariance Network Dysfunction With Neuropsychiatric Symptoms Across the Alzheimer's Disease Continuum. Biol Psychiatry 2025; 97:1067-1078. [PMID: 39419461 DOI: 10.1016/j.biopsych.2024.08.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 07/04/2024] [Accepted: 08/28/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND The high heterogeneity of neuropsychiatric symptoms (NPSs) hinders further exploration of their role in neurobiological mechanisms and Alzheimer's disease (AD). We aimed to delineate NPS patterns based on brain macroscale connectomics to understand the biological mechanisms of NPSs on the AD continuum. METHODS We constructed regional radiomics similarity networks for 550 participants (AD with NPSs [n = 376], AD without NPSs [n = 111], and normal control participants [n = 63]) from the CIBL (Chinese Imaging, Biomarkers, and Lifestyle) study. We identified regional radiomics similarity network connections associated with NPSs and then clustered distinct subtypes of AD with NPSs. An independent dataset (n = 189) and internal validation were performed to assess the robustness of the NPS subtypes. Subsequent multiomics analysis was performed to assess the distinct clinical phenotype and biological mechanisms in each NPS subtype. RESULTS AD patients with NPSs were clustered into severe (n = 187), moderate (n = 87), and mild (n = 102) NPS subtypes, each exhibiting distinct brain network dysfunction patterns. A high level of consistency in clustering NPSs was internally and externally validated. Severe and moderate NPS subtypes were associated with significant cognitive impairment, increased plasma p-tau181 (tau phosphorylated at threonine 181) levels, extensive decreased brain volume and cortical thickness, and accelerated cognitive decline. Gene set enrichment analysis revealed enrichment of differentially expressed genes in ion transport and synaptic transmission with variations for each NPS subtype. Genome-wide association study analysis defined the specific gene loci for each subtype of AD with NPSs (e.g., logical memory), consistent with clinical manifestations and progression patterns. CONCLUSIONS This study identified and validated 3 distinct NPS subtypes, underscoring the role of NPSs in neurobiological mechanisms and progression of the AD continuum.
Collapse
Affiliation(s)
- Jiwei Jiang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Kun Zhao
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China; Queen Mary School Hainan, Beijing University of Posts and Telecommunications, Hainan, China.
| | - Wenyi Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Peiyang Zheng
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
| | - Shirui Jiang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Qiwei Ren
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yunyun Duan
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Huiying Yu
- School of Information Science and Engineering, Shandong Normal University, Jinan, China
| | - Xiaopeng Kang
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Junjie Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ke Hu
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Tianlin Jiang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Min Zhao
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Linlin Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Shiyi Yang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Huiying Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yaou Liu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Anxin Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yong Liu
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China; Queen Mary School Hainan, Beijing University of Posts and Telecommunications, Hainan, China.
| | - Jun Xu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China.
| |
Collapse
|
14
|
Wang X, Qiao Y, Liang Y, Han J, Duan H, Tang Y, Qin Q. Incidence and Risk Factors of Post-Lumbar Puncture Headache in Patients With Cognitive Impairment. Brain Behav 2025; 15:e70597. [PMID: 40444538 PMCID: PMC12123446 DOI: 10.1002/brb3.70597] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 05/07/2025] [Accepted: 05/09/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND Cognitive impairment is prevalent among the elderly population. Cerebrospinal fluid (CSF) biomarker tests are employed to facilitate timely and differential diagnosis of cognitive dysfunction. OBJECTIVE To investigate incidence and risk factors of post-lumbar puncture headache (PLPH) in Chinese geriatric patients with cognitive impairment. METHODS A total of 254 consecutive patients with cognitive impairment were recruited between March and September 2023 and categorized into two groups: mild cognitive impairment (MCI) and dementia. Different scales, such as the short-form of the McGill pain questionnaire (SF-MPQ), facial visual analogue scale (F-VAS), revised Wong-Baker Assessment of Facial Expression Pain (FPS-R), and Chinese version of Pain Assessment Scale for Advanced Dementia (C-PAINAD), were used to evaluate the incidence of headache after lumbar puncture. Univariate and multivariate factor analyses were conducted to identify potential risk factors, with the most influential predictors included in regression models. RESULTS Among patients with cognitive disorders, the incidence of PLPH was 24.8%. The incidence and duration of headache did not differ significantly between the MCI and dementia groups. The average time of post-lumbar puncture headache (PLPH) was 22.9 h. There was no significant difference in headache scale scores between MCI patients. The incidence of PLPH in women with cognitive impairment was higher than that in men. The incidence of PLPH in cognitive impairment patients with a history of headache was significantly higher than that in cognitive impairment patients without a history of headache. However, no significant differences were observed in years of education, number of lumbar punctures, length of bed rest, and oral rehydration volume. CONCLUSION PLPH is more commonly observed in women, individuals with a history of headache, younger age, or lower body mass index (BMI). C-PAINAD and SF-MPQ are the preferred assessment tools for evaluating PLPH in patients with cognitive impairment.
Collapse
Affiliation(s)
- Xinhui Wang
- Department of GeriatricsHenan Provincial People's Hospital, People's Hospital of Zhengzhou UniversityZhengzhouChina
- Innovation Center for Neurological Disorders, Department of NeurologyXuanwu Hospital, Capital Medical University, National Center for Neurological DisordersBeijingChina
| | - Yuchen Qiao
- Innovation Center for Neurological Disorders, Department of NeurologyXuanwu Hospital, Capital Medical University, National Center for Neurological DisordersBeijingChina
| | | | - Jinming Han
- Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Hongyan Duan
- Department of GeriatricsHenan Provincial People's Hospital, People's Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yi Tang
- Innovation Center for Neurological Disorders, Department of NeurologyXuanwu Hospital, Capital Medical University, National Center for Neurological DisordersBeijingChina
| | - Qi Qin
- Innovation Center for Neurological Disorders, Department of NeurologyXuanwu Hospital, Capital Medical University, National Center for Neurological DisordersBeijingChina
| |
Collapse
|
15
|
Li Q, Cui L, Guan Y, Li Y, Xie F, Guo Q. Prediction Model and Nomogram for Amyloid Positivity Using Clinical and MRI Features in Individuals With Subjective Cognitive Decline. Hum Brain Mapp 2025; 46:e70238. [PMID: 40439500 PMCID: PMC12121204 DOI: 10.1002/hbm.70238] [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: 11/20/2024] [Revised: 04/10/2025] [Accepted: 05/17/2025] [Indexed: 06/02/2025] Open
Abstract
There is an urgent need for the precise prediction of cerebral amyloidosis using noninvasive and accessible indicators to facilitate the early diagnosis of individuals with the preclinical stage of Alzheimer's disease (AD). Two hundred and four individuals with subjective cognitive decline (SCD) were enrolled in this study. All subjects completed neuropsychological assessments and underwent 18F-florbetapir PET, structural MRI, and functional MRI. A total of 315 features were extracted from the MRI, demographics, and neuropsychological scales and selected using the least absolute shrinkage and selection operator (LASSO). The logistic regression (LR) model, based on machine learning, was trained to classify SCD as either β-amyloid (Aβ) positive or negative. A nomogram was established using a multivariate LR model to predict the risk of Aβ+. The performance of the prediction model and nomogram was assessed with area under the curve (AUC) and calibration. The final model was based on the right rostral anterior cingulate thickness, the grey matter volume of the right inferior temporal, the ReHo of the left posterior cingulate gyrus and right superior temporal gyrus, as well as MoCA-B and AVLT-R. In the training set, the model achieved a good AUC of 0.78 for predicting Aβ+, with an accuracy of 0.72. The validation of the model also yielded a favorable discriminatory ability with an AUC of 0.88 and an accuracy of 0.83. We have established and validated a model based on cognitive, sMRI, and fMRI data that exhibits adequate discrimination. This model has the potential to predict amyloid status in the SCD group and provide a noninvasive, cost-effective way that might facilitate early screening, clinical diagnosis, and drug clinical trials.
Collapse
Affiliation(s)
- Qinjie Li
- Department of GerontologyShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Liang Cui
- Department of GerontologyShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Yihui Guan
- PET Center, Huashan HospitalFudan UniversityShanghaiChina
| | - Yuehua Li
- Department of RadiologyShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Fang Xie
- PET Center, Huashan HospitalFudan UniversityShanghaiChina
| | - Qihao Guo
- Department of GerontologyShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| |
Collapse
|
16
|
Cao Y, Xu Y, Cao M, Chen N, Zeng Q, Lai MKP, Fan D, Sethi G, Cao Y. Fluid-based biomarkers for neurodegenerative diseases. Ageing Res Rev 2025; 108:102739. [PMID: 40122396 DOI: 10.1016/j.arr.2025.102739] [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: 09/01/2024] [Revised: 03/10/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Neurodegenerative diseases, such as Alzheimer's Disease (AD), Multiple Sclerosis (MS), Parkinson's Disease (PD), and Amyotrophic Lateral Sclerosis (ALS) are increasingly prevalent as global populations age. Fluid biomarkers, derived from cerebrospinal fluid (CSF), blood, saliva, urine, and exosomes, offer a promising solution for early diagnosis, prognosis, and disease monitoring. These biomarkers can reflect critical pathological processes like amyloid-beta (Aβ) deposition, tau protein hyperphosphorylation, α-syn misfolding, TDP-43 mislocalization and aggregation, and neuronal damage, enabling detection long before clinical symptoms emerge. Recent advances in blood-based biomarkers, particularly plasma Aβ, phosphorylated tau, and TDP-43, have shown diagnostic accuracy equivalent to CSF biomarkers, offering more accessible testing options. This review discusses the current challenges in fluid biomarker research, including variability, standardization, and sensitivity issues, and explores how combining multiple biomarkers with clinical symptoms improves diagnostic reliability. Ethical considerations, future directions involving extracellular vehicles (EVs), and the integration of artificial intelligence (AI) are also highlighted. Continued research efforts will be key to overcoming these obstacles, enabling fluid biomarkers to become crucial tools in personalized medicine for neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Yifei Xu
- Institute of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, China
| | - Meiqun Cao
- Department of Neurology, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China
| | - Nan Chen
- Institute of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, China
| | - Qingling Zeng
- Institute of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, China
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, 117600, Singapore; Memory, Aging and Cognition Centre, National University Health System, Singapore
| | - Dahua Fan
- Institute of Maternal-Fetal Medicine,Shunde Women and Children's Hospital, Guangdong Medical University, Foshan 528300, China.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, 117600, Singapore; NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore.
| | - Yongkai Cao
- Department of Neurology, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen 518035, China.
| |
Collapse
|
17
|
Kicik A, Kurt E, Hari E, Ulasoglu-Yildiz Ç, Gurvit H, Demiralp T. Impact of amygdala functional connectivity on cognitive impairment in Alzheimer's disease. Neurol Sci 2025; 46:2601-2610. [PMID: 40055296 DOI: 10.1007/s10072-025-08091-0] [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: 12/08/2024] [Accepted: 02/27/2025] [Indexed: 05/17/2025]
Abstract
The functional connectivity (FC) of the amygdala in Alzheimer's disease (AD) and its relationship to cognitive impairment is still not well established. Thus, we examined resting-state FC changes in the amygdala among 21 patients with AD dementia (ADD) and 34 individuals with amnestic mild cognitive impairment (aMCI), compared to 33 individuals with subjective cognitive impairment (SCI), to provide insights into the association between amygdala FC and cognitive decline in different clinical stages of Alzheimer's disease. We conducted seed-to-voxel FC analysis, focused on two cognitive functions, episodic memory, and face recognition, and examined the correlations between changes in FC of the amygdala and cognitive test scores. We demonstrated that the left amygdala exhibits progressive disruption in FC, especially with the frontal regions in aMCI and ADD. We further identified that this disrupted FC in the left amygdala showed significant positive correlations with cognitive test scores from the MCI stage onward. Our results indicate that FC changes in the left amygdala may serve as an early marker of AD and this FC pattern of amygdala influence detrimentally affects episodic memory and face recognition functions. These findings highlight that the amygdala may be a critical anatomical region for detecting the early stages of AD.
Collapse
Affiliation(s)
- Ani Kicik
- Department of Physiology, Faculty of Medicine, Demiroglu Bilim University, Istanbul, 34394, Turkey.
| | - Elif Kurt
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, 34093, Turkey
| | - Emre Hari
- Department of Physiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, 34093, Turkey
- Graduate School of Health Sciences, Istanbul University, Istanbul, 34216, Turkey
| | - Çiğdem Ulasoglu-Yildiz
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, 34093, Turkey
| | - Hakan Gurvit
- Behavioral Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, 34093, Turkey
| | - Tamer Demiralp
- Department of Physiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, 34093, Turkey
- Hulusi Behcet Life Sciences Research Laboratory, Neuroimaging Unit, Istanbul University, Istanbul, 34093, Turkey
| |
Collapse
|
18
|
Blanco MN, Szpiro AA, Crane PK, Sheppard L. Ultrafine particles and late-life cognitive function: Influence of stationary mobile monitoring design on health inferences. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 374:126222. [PMID: 40221115 PMCID: PMC12050196 DOI: 10.1016/j.envpol.2025.126222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
Growing evidence links ultrafine particles (UFP) to neurotoxicity, but human studies remain limited. Various mobile monitoring approaches have been used to develop air pollution exposure models. However, whether design choices impact epidemiology, including for UFP and cognitive function, remains unclear. We evaluated the adjusted association between 5-year average UFP number concentration (PNC) and late-life cognitive function (Cognitive Abilities Screening Instrument - Item Response Theory [CASI-IRT]) in the Adult Changes in Thought cohort (N = 5283) by leveraging an extensive roadside mobile monitoring campaign specifically designed for epidemiology. To assess the impact of reduced monitoring approaches on this association, we repeatedly subsampled UFP measures from the campaign, developed exposure models, and evaluated the degree to which associations were impacted. In the primary analysis, each 1900 pt/cm3 increment in PNC was associated with an adjusted mean baseline CASI-IRT score that was 0.002 (95 % CI: -0.016, 0.020) higher, which was not statistically significant. Point estimates were consistent across sampling designs with fewer visits per site (≤12), fewer seasons (1-3), and unbalanced visit frequency across sites. Sampling designs restricted to rush hours were more similar (median point estimate 0.002, IQR of point estimates: 0.000, 0.003) than business hour designs (0.006, IQR: 0.005, 0.007), but the opposite was true when temporal adjustments were applied (rush: -0.003, IQR: -0.005, -0.001; business: 0.002, IQR: 0.001, 0.004). We observed similar results in sensitivity and secondary analyses. We did not find evidence of an association between UFP and cognitive function in fully adjusted models. Monitoring design had minimal impact on the inferential results in this setting, which may have been caused by the lack of association. Secondary analyses in a reduced model that is potentially confounded suggest that monitoring design might have a greater impact in other datasets. Further research is needed, particularly in contexts with robust statistically significant health associations.
Collapse
Affiliation(s)
- Magali N Blanco
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA.
| | - Adam A Szpiro
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Paul K Crane
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Lianne Sheppard
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA; Department of Biostatistics, University of Washington, Seattle, WA, USA
| |
Collapse
|
19
|
Chen L, Zhou X, Qiao Y, Wang Y, Zhou Z, Jia S, Sun Y, Peng D. The impact of Alzheimer's disease on cortical complexity and its underlying biological mechanisms. Brain Res Bull 2025; 225:111320. [PMID: 40189107 DOI: 10.1016/j.brainresbull.2025.111320] [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: 11/21/2024] [Revised: 03/07/2025] [Accepted: 03/24/2025] [Indexed: 04/17/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) might impact the complexity of cerebral cortex, and the underlying biological mechanisms responsible for cortical changes in the AD cortex remain unclear. METHODS Fifty-eight participants with AD and 67 normal controls underwent high-resolution 3 T structural brain MRI. Using surface-based morphometry (SBM), we created vertex-wise maps for group comparisons in terms of five measures: cortical thickness, fractal dimension, gyrification index, Toro's gyrification index and sulcal depth respectively. Five machine learning (ML) models combining SBM parameters were established to predict AD. In addition, transcription-neuroimaging association analyses, as well as Mendelian randomization of AD and cortical thickness data, were conducted to investigate the genetic mechanisms and biological functions of AD. RESULTS AD patients exhibited topological changes in cortical complexity, with increased complexity in the frontal and temporal cortex and decreased complexity in the insula cortex, alongside extensive cortical atrophy. Combining different SBM measures could aid disease diagnosis. The genes involved in cell structure support and the immune response were the strongest contributors to cortical anatomical features in AD patients. The identified genes associated with AD cortical morphology were overexpressed or underexpressed in excitatory neurons, oligodendrocytes, and astrocytes. CONCLUSION Complexity alterations of the cerebral surface may be associated with a range of biological processes and molecular mechanisms, including immune responses. The present findings may contribute to a more comprehensive understanding of brain morphological patterns in AD patients.
Collapse
Affiliation(s)
- Leian Chen
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Xiao Zhou
- Department of Neurology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yanan Qiao
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Yu Wang
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Zhi Zhou
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Shuhong Jia
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China
| | - Yu Sun
- Department of Neurology, China-Japan Friendship Hospital, Beijing, China.
| | - Dantao Peng
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Department of Neurology, China-Japan Friendship Hospital, Beijing, China.
| |
Collapse
|
20
|
van der Meer A, Knippenberg M, Visser D, Boenink M. Anticipating the Future in an Uncertain Present: How Individuals Living With Medically Unexplained Cognitive Complaints Value Biomarker-Based Prognostics. Health Expect 2025; 28:e70284. [PMID: 40411282 PMCID: PMC12102610 DOI: 10.1111/hex.70284] [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: 06/15/2024] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 05/26/2025] Open
Abstract
INTRODUCTION In research on Alzheimer's disease (AD) individuals experiencing cognitive problems but not satisfying criteria for mild cognitive impairment (MCI) or AD are referred to as having 'Subjective Cognitive Decline' (SCD). A small subset of them will progress to a state justifying an AD diagnosis. AD research trying to identify prognostic biomarkers aims to inform individuals labelled with SCD about the future development of their complaints. This paper explores how persons experiencing these cognitive complaints currently anticipate the future and how they would value emerging biomarker-based prognostic tests. METHODS Semi-structured interviews with 10 individuals experiencing cognitive complaints, some accompanied by their partners. The interviews were coded using ATLAS.ti 22. The exploratory qualitative analysis was conducted using thematic content analysis (TA). FINDINGS The interviewees experience pervasive uncertainty concerning their persistent but medically unexplained cognitive symptoms. Lacking a diagnosis and a prognosis, they anticipate their future considering various scenarios, while mainly acting upon the scenario of developing further cognitive decline/AD dementia. Interviewees often interpret questions about prognostication as asking about predicting the likelihood of a future diagnosis, clearly valuing the latter. Most interviewees are positive, assuming it would help them and their relatives prepare for the future. They have specific ideas about what type of prognostic information would be helpful, usually focusing on how cognitive decline would impact their lives, for example, what activities they will be able to continue to engage in, when they will no longer recognise loved ones or what character they will have as a patient. CONCLUSION People experiencing unexplained cognitive complaints experience pervasive uncertainty about their situation and future. They clearly value the idea of a test predicting with high certainty whether they will be diagnosed with AD in the future. Regarding prognostication, most interviewees expect that such tests could help prepare for the future. However, there is a discrepancy between the outcomes usually measured in prognostic research (like the expected speed of decline) and the specific outcomes meaningful to our interviewees. In view of this gap, it is important to reconsider whether and how the development of prognostic biomarker tests for people suffering from unexplained cognitive complaints can be better tailored to the needs of these individuals. PATIENT OR PUBLIC CONTRIBUTION During our interviews, we gathered the perspectives of 10 individuals suffering from medically unexplained cognitive complaints on the value of biomarker-based prognostics. These interviews were the primary data source. Individuals were also involved in the interpretation of our findings and critical evaluation of our preliminary conclusions.
Collapse
Affiliation(s)
- Anne‐Fleur van der Meer
- Radboud University Medical Centre, IQ Health, Ethics of Healthcare GroupNijmegenThe Netherlands
| | - Marjan Knippenberg
- Radboud University Medical Centre, IQ Health, Ethics of Healthcare GroupNijmegenThe Netherlands
| | - Denise Visser
- Department of Radiology & Nuclear Medicine, Amsterdam UMCVrije Universiteit AmsterdamAmsterdamThe Netherlands
- Amsterdam Neuroscience, Brain ImagingAmsterdamThe Netherlands
| | - Marianne Boenink
- Radboud University Medical Centre, IQ Health, Ethics of Healthcare GroupNijmegenThe Netherlands
| |
Collapse
|
21
|
Qiang Q, Skudder-Hill L, Toyota T, Huang Z, Wei W, Adachi H. CSF α-synuclein aggregation is associated with APOE ε4 and progressive cognitive decline in Alzheimer's disease. Neurobiol Aging 2025; 150:9-18. [PMID: 40043469 DOI: 10.1016/j.neurobiolaging.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/23/2025] [Accepted: 02/26/2025] [Indexed: 04/10/2025]
Abstract
At autopsy, around half of the Alzheimer's disease (AD) brains exhibit Lewy body pathology, and the main component of Lewy body pathology is α-synuclein aggregates. This study investigated the prevalence of cerebrospinal fluid (CSF) α-synuclein aggregation and its association with demographic factors and cognitive decline among 1619 participants from the Alzheimer's Disease Neuroimaging Initiative (ADNI), with the test for α-synuclein aggregation by seed amplification assay (SAA). This cohort consisted of 595 cognitively normal (CN) individuals, 765 with mild cognitive impairment (MCI), and 259 with AD dementia. The results showed a higher prevalence of positive α-synuclein aggregation status in the AD dementia group (37.07 %) and the MCI group (22.75 %) compared to CN controls (16.13 %). Additionally, APOE ε4 carriers exhibited a higher prevalence of α-synuclein aggregation compared to non-carriers: 20.12 % for APOE ε4-/- (non-carriers), 24.82 % for APOE ε4 + /-, and 30.92 % for APOE ε4 + /+ . Longitudinally, positive CSF α-synuclein aggregation associated with accelerated cognitive decline, especially in the MCI and AD groups. Notably, positive aggregation status did not significantly affect cognitive trajectories in CN individuals. Moreover, APOE ε4 carriers with positive CSF α-synuclein aggregation experienced more pronounced cognitive decline. This study provides evidence that CSF α-synuclein aggregation is associated with cognitive function and the APOE ε4 allele. These findings suggest that CSF α-synuclein SAA, in combination with APOE ε4 status, could serve as biomarkers for predicting cognitive decline in AD.
Collapse
Affiliation(s)
- Qiang Qiang
- Department of Neurology, Cognitive Disorders Center, Huadong Hospital, Fudan University, Shanghai, China; Department of Neurology, University of Occupational and Environmental Health School of Medicine, Kitakyushu, Japan
| | - Loren Skudder-Hill
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Tomoko Toyota
- Department of Neurology, University of Occupational and Environmental Health School of Medicine, Kitakyushu, Japan
| | - Zhe Huang
- Department of Neurology, University of Occupational and Environmental Health School of Medicine, Kitakyushu, Japan
| | - Wenshi Wei
- Department of Neurology, Cognitive Disorders Center, Huadong Hospital, Fudan University, Shanghai, China
| | - Hiroaki Adachi
- Department of Neurology, University of Occupational and Environmental Health School of Medicine, Kitakyushu, Japan.
| |
Collapse
|
22
|
Galvin JE, Kleiman MJ, Harris HM, Estes PW. The Cognivue Amyloid Risk Measure (CARM): A Novel Method to Predict the Presence of Amyloid with Cognivue Clarity. Neurol Ther 2025; 14:865-880. [PMID: 40192926 PMCID: PMC12089551 DOI: 10.1007/s40120-025-00741-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 03/26/2025] [Indexed: 05/20/2025] Open
Abstract
INTRODUCTION At the present time, clinical detection of individuals who have amyloid in their brain is not possible without expensive biomarkers. The objective of the study was to test whether Cognivue Clarity® can differentiate True Controls, preclinical Alzheimer's disease (pAD), mild cognitive impairment (MCI) due to Alzheimer's disease (MCI-AD), AD, and MCI and dementia due to non-AD etiologies enrolled in the Bio-Hermes Study. METHODS A total of 887 individuals completed Cognivue Clarity, amyloid PET scan, and blood-based AD biomarkers. Three Cognivue Clarity subtests differentiated between True Controls and pAD, and between cognitive impairment due to AD versus non-AD processes. This finding was leveraged to develop an amyloid-specific marker, combining the three subtests with age using machine learning to create the 4-point Cognivue Amyloid Risk Measure (CARM). RESULTS Cognivue Clarity discriminated cognitively normal from cognitively impaired individuals (p < 0.001, Cohen's d = 0.732). The CARM differentiated between individuals with amyloid and without amyloid by PET (p < 0.001, Cohen's d = 0.618) and blood-based biomarkers (p's < 0.001). Amyloid positivity and cognitive impairment increased across four CARM thresholds (p < 0.001). Dichotomizing CARM thresholds into low (CARM1/CARM2) and high (CARM3/CARM4) likelihood provided excellent discrimination for amyloid PET positivity (OR: 3.67; 95% CI 2.76-4.89). CARM categories differentiated between True Controls, pAD, MCI-AD, AD, and cognitive impairment due to non-AD etiologies (χ2 = 137.6, p < 0.001) with the majority of True Controls and non-AD etiologies being in CARM1/CARM2, and the majority of pAD, MCI-AD, and AD being in CARM3/CARM4. CONCLUSIONS Cognivue Clarity detects individuals with cognitive impairment, and a derivation benchmarked against amyloid PET was used to develop the CARM to predict the presence of amyloid. Combining the CARM and the Cognivue Clarity overall score could help identify individuals with and without cognitive impairment due to AD or non-AD etiologies, help screen for treatment protocols with anti-amyloid therapies, enrich clinical trial recruitment, and help to identify pAD for prevention studies. TRIAL REGISTRATION ClinicalTrials. gov identifier, NCT04733989.
Collapse
Affiliation(s)
- James E Galvin
- Department of Neurology, Comprehensive Center for Brain Health, University of Miami Miller School of Medicine, 7700 W Camino Real, Suite 200, Boca Raton, FL, 33433, USA.
- Cognivue, Inc, 7911 Rae Blvd, Victor, NY, 14564, USA.
| | - Michael J Kleiman
- Department of Neurology, Comprehensive Center for Brain Health, University of Miami Miller School of Medicine, 7700 W Camino Real, Suite 200, Boca Raton, FL, 33433, USA
| | | | - Paul W Estes
- Cognivue, Inc, 7911 Rae Blvd, Victor, NY, 14564, USA
| |
Collapse
|
23
|
Zhang W, Sun C, Huang Y, Zhang M, Xu A, Wang C, Lv F, Pan T. Inflammation levels in type 2 diabetes mellitus patients with mild cognitive impairment: Assessment followed by amelioration via dapagliflozin therapy. J Diabetes Complications 2025; 39:109017. [PMID: 40228375 DOI: 10.1016/j.jdiacomp.2025.109017] [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: 03/03/2025] [Revised: 03/21/2025] [Accepted: 03/22/2025] [Indexed: 04/16/2025]
Abstract
AIMS To investigate systemic inflammation and the effect of dapagliflozin treatment in (type 2 diabetes mellitus) T2DM patients with mild cognitive impairment (MCI). METHODS Between January and December 2023, 200 participants were recruited from the Department of Endocrinology of Hefei First People's Hospital. Baseline data collected included medical history, fasting blood glucose, HbA1c, liver and kidney function, lipid profiles, IL-1β, TNF-α, sVCAM-1 level, and the urinary albumin-creatinine ratio (uACR). Based on their Montreal Cognitive Assessment Scale (MoCA) scores, these participants were categorized into two groups: 127 in the MCI group and 73 in the non-MCI group. MCI group received dapagliflozin (10 mg daily) alongside standard treatment. RESULTS The MCI group showed higher age, height, weight, BMI, HbA1c, FBG, disease duration, carotid plaques, stenosis rates, and elevated IL-1β, TNF-α, and sVCAM-1. MoCA scores were significantly lower in the MCI group. Correlation analysis showed a negative correlation of MoCA scores with IL-1β, TNF-α, sVCAM-1, plaques, stenosis, FBG, and HbA1c, and a positive correlation with height. Binary logistic regression identified age, BMI, IL-1β, sVCAM-1, and FBG as predictors of cognitive impairment in T2DM. Dapagliflozin treatment reduced BMI, HbA1c, inflammatory markers, and FBG, improving MoCA scores. CONCLUSION Dapagliflozin treatment may improve cognitive function by reducing inflammation.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Endocrinology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Department of Endocrinology, The Third Affiliated Hospital of Anhui Medical University (Hefei First People's Hospital), Hefei 230061, China
| | - Chunping Sun
- Department of Endocrinology, The Third Affiliated Hospital of Anhui Medical University (Hefei First People's Hospital), Hefei 230061, China
| | - Yating Huang
- Department of Endocrinology, The Third Affiliated Hospital of Anhui Medical University (Hefei First People's Hospital), Hefei 230061, China
| | - Meng Zhang
- Department of Endocrinology, The Third Affiliated Hospital of Anhui Medical University (Hefei First People's Hospital), Hefei 230061, China
| | - Ao Xu
- Department of Endocrinology, The Third Affiliated Hospital of Anhui Medical University (Hefei First People's Hospital), Hefei 230061, China
| | - Chen Wang
- Department of Endocrinology, The Third Affiliated Hospital of Anhui Medical University (Hefei First People's Hospital), Hefei 230061, China
| | - Fang Lv
- Department of Endocrinology, The Third Affiliated Hospital of Anhui Medical University (Hefei First People's Hospital), Hefei 230061, China.
| | - Tianrong Pan
- Department of Endocrinology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China.
| |
Collapse
|
24
|
Bhatia V, Chandel A, Minhas Y, Kushawaha SK. "Advances in biomarker discovery and diagnostics for alzheimer's disease". Neurol Sci 2025; 46:2419-2436. [PMID: 39893357 DOI: 10.1007/s10072-025-08023-y] [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: 11/23/2024] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by intracellular neurofibrillary tangles with tau protein and extracellular β-amyloid plaques. Early and accurate diagnosis is crucial for effective treatment and management. OBJECTIVE The purpose of this review is to investigate new technologies that improve diagnostic accuracy while looking at the current diagnostic criteria for AD, such as clinical evaluations, cognitive testing, and biomarker-based techniques. METHODS A thorough review of the literature was done in order to assess both conventional and contemporary diagnostic methods. Multimodal strategies integrating clinical, imaging, and biochemical evaluations were emphasised. The promise of current developments in biomarker discovery was also examined, including mass spectrometry and artificial intelligence. RESULTS Current diagnostic approaches include cerebrospinal fluid (CSF) biomarkers, imaging tools (MRI, PET), cognitive tests, and new blood-based markers. Integrating these technologies into multimodal diagnostic procedures enhances diagnostic accuracy and distinguishes dementia from other conditions. New technologies that hold promise for improving biomarker identification and diagnostic reliability include mass spectrometry and artificial intelligence. CONCLUSION Advancements in AD diagnostics underscore the need for accessible, minimally invasive, and cost-effective techniques to facilitate early detection and intervention. The integration of novel technologies with traditional methods may significantly enhance the accuracy and feasibility of AD diagnosis.
Collapse
Affiliation(s)
- Vandana Bhatia
- Department of Pharmacology, Laureate Institute of Pharmacy Kathog, Kangra, 177101, India.
| | - Anjali Chandel
- Department of Pharmacology, Laureate Institute of Pharmacy Kathog, Kangra, 177101, India
| | - Yavnika Minhas
- Department of Pharmacology, Laureate Institute of Pharmacy Kathog, Kangra, 177101, India
| | | |
Collapse
|
25
|
Zhao K, Chen P, Wang D, Zhou R, Ma G, Liu Y. A Multiform Heterogeneity Framework for Alzheimer's Disease Based on Multimodal Neuroimaging. Biol Psychiatry 2025; 97:1022-1033. [PMID: 39725298 DOI: 10.1016/j.biopsych.2024.12.009] [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: 09/19/2024] [Revised: 11/14/2024] [Accepted: 12/15/2024] [Indexed: 12/28/2024]
Abstract
Understanding the heterogeneity of Alzheimer's disease (AD) is crucial for advancing precision medicine specifically tailored to this disorder. Recent research has deepened our understanding of AD heterogeneity; however, translating these insights from bench to bedside via neuroimaging heterogeneity frameworks presents significant challenges. In this review, we systematically revisit prior studies and summarize the existing methodology of data-driven neuroimaging studies for AD heterogeneity. We organized the current methodology into 1) a subtyping clustering strategy for patients with AD, and we also subdivided it into subtyping analysis based on cross-sectional multimodal neuroimaging profiles and the identification of long-term disease progression from short-term datasets; 2) a stratified strategy that integrates neuroimaging measures with biomarkers; and 3) individual-specific abnormal patterns based on the normative model. Then, we evaluated the characteristics of these studies along 2 dimensions: 1) the understanding of pathology and 2) clinical application. We systematically address the limitations, challenges, and future directions of research into AD heterogeneity. Our goal is to enhance the neuroimaging heterogeneity framework for AD, thereby facilitating its transition from bench to bedside.
Collapse
Affiliation(s)
- Kun Zhao
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China; Queen Mary School Hainan, Beijing University of Posts and Telecommunications, Hainan, China
| | - Pindong Chen
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Dong Wang
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China
| | - Rongshen Zhou
- School of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Guolin Ma
- Department of Radiology, China-Japan Friendship Hospital, Beijing, China
| | - Yong Liu
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, China; Queen Mary School Hainan, Beijing University of Posts and Telecommunications, Hainan, China; Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing, China; Center for Inspur-BUPT, Beijing University of Posts and Telecommunications, Beijing, China.
| |
Collapse
|
26
|
Rullmann M, Henssen D, Melasch JT, Scherlach C, Saur D, Schroeter ML, Tiepolt S, Koglin N, Stephens AW, Hesse S, Strauss M, Brendel M, Mishchenko O, Schildan A, Classen J, Hoffmann KT, Sabri O, Barthel H, German Imaging Initiative for Tauopathies (GII4T). Multi-parametric [ 18F]PI-2620 tau PET/MRI for the phenotyping of different Alzheimer's disease variants. Eur J Nucl Med Mol Imaging 2025; 52:2279-2289. [PMID: 39937274 PMCID: PMC12119744 DOI: 10.1007/s00259-025-07135-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 01/31/2025] [Indexed: 02/13/2025]
Abstract
PURPOSE Heterogeneity in clinical phenotypes has led to the description of different phenotypes of Alzheimer's disease (AD). Besides the most frequent amnestic variant of AD (aAD), patients presenting with language deficits are diagnosed with logopenic variant primary progressive aphasia (lvPPA), whereas patients presenting with visual deficits are classified as posterior cortical atrophy (PCA). METHODS This study set out to investigate the value of a multi-parametric [18F]PI-2620 tau PET/MRI protocol to distinguish aAD, lvPPA and PCA to support clinical diagnosis in 32 patients. Phenotype-specific information about tau accumulation, relative perfusion, grey matter density, functional network alterations and white matter microstructural alterations was collected. RESULTS The aAD patients showed significantly higher tau accumulation, relative hypoperfusion and grey matter density loss in the temporal lobes compared to PCA and lvPPA patients. PCA patients, on the other hand, showed significantly higher tau accumulation in the occipital lobe as compared to aAD patients. Relative hypoperfusion in the occipital lobe and loss of functional connectivity of the posterior cingulate cortex to supplementary visual cortical regions helped to distinguish PCA from lvPPA. Tau accumulation in the cerebellum and microstructural changes in the cingulum were found to help differentiate lvPPA from aAD. CONCLUSION This study highlights structural and functional differences between patients with different AD phenotypes. Differences in regional tau PET signals suggest that refinements in the Braak staging system are needed for the non-aAD cases. These patterns of tau accumulation align with the cascading network failure hypothesis, though more research is needed to warrant the here presented results in larger patient cohorts.
Collapse
Affiliation(s)
- Michael Rullmann
- Department of Nuclear Medicine, University of Leipzig Medical Center Leipzig, Leipzig, Germany.
| | - Dylan Henssen
- Department of Nuclear Medicine, University of Leipzig Medical Center Leipzig, Leipzig, Germany
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Juliana T Melasch
- Department of Nuclear Medicine, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Cordula Scherlach
- Department of Neuroradiology, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Dorothee Saur
- Department of Neurology, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Matthias L Schroeter
- Clinic for Cognitive Neurology, University of Leipzig Medical Center Leipzig, Leipzig, Germany
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Solveig Tiepolt
- Department of Nuclear Medicine, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | | | | | - Swen Hesse
- Department of Nuclear Medicine, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Maria Strauss
- Department of Psychiatry, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Olena Mishchenko
- Department of Nuclear Medicine, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Andreas Schildan
- Department of Nuclear Medicine, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Joseph Classen
- Department of Neurology, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Karl-Titus Hoffmann
- Department of Neuroradiology, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, University of Leipzig Medical Center Leipzig, Leipzig, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University of Leipzig Medical Center Leipzig, Leipzig, Germany
- Department of Nuclear Medicine, Hospital Dessau, Dessau, Germany
| | | |
Collapse
|
27
|
Heneka MT, Gauthier S, Chandekar SA, Hviid Hahn-Pedersen J, Bentsen MA, Zetterberg H. Neuroinflammatory fluid biomarkers in patients with Alzheimer's disease: a systematic literature review. Mol Psychiatry 2025; 30:2783-2798. [PMID: 40050444 PMCID: PMC12092255 DOI: 10.1038/s41380-025-02939-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 01/08/2025] [Accepted: 02/19/2025] [Indexed: 05/22/2025]
Abstract
INTRODUCTION Neuroinflammation is associated with both early and late stages of the pathophysiology of Alzheimer's disease (AD). Fluid biomarkers are gaining significance in clinical practice for diagnosis in presymptomatic stages, monitoring, and disease prognosis. This systematic literature review (SLR) aimed to identify fluid biomarkers for neuroinflammation related to clinical stages across the AD continuum and examined long-term outcomes associated with changes in biomarkers. METHODS The SLR was conducted per the Cochrane Handbook for Systematic Reviews of Interventions and Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. We used PubMed®, Embase®, and Cochrane Collaboration databases to search for articles in English (between 2012 and 2022) on AD or mild cognitive impairment due to AD, using "neuroinflammation" or other "immune" search strings. Two independent reviewers screened titles and examined data from full-text articles for the SLR. RESULTS After the initial screening, 54 studies were prioritized for data extraction based upon their relevance to the SLR research questions. Nine studies for YKL-40, seven studies for sTREM2, and 11 studies for GFAP examined the relationship between the neuroinflammatory biomarkers and the clinical stage of the disease. Nine longitudinal studies further explored the association of fluid biomarkers with long-term clinical outcomes of disease. Cerebrospinal fluid (CSF) levels of YKL-40 were elevated in patients with AD dementia, while CSF sTREM2 levels were more strongly associated with preclinical and early symptomatic stages of AD. Plasma GFAP levels remained consistently elevated both in patients with AD dementia and individuals in preclinical stages with β-amyloid pathology. Longitudinal changes in plasma GFAP appeared to be predictive of cognitive decline in patients over time. DISCUSSION Neuroinflammatory biomarkers are associated with AD progression. More longitudinal studies in the preclinical and MCI stages of AD are needed to validate fluid biomarkers for diagnosis, disease monitoring, and prognosis in clinical practice.
Collapse
Affiliation(s)
- Michael T Heneka
- Luxembourg Centre for Systems Biomedicine, Université du Luxembourg, Belvaux, Luxembourg.
| | - Serge Gauthier
- AD and Related Disorders Research Unit, McGill Center for Studies in Aging, Departments of Neurology & Neurosurgery, Psychiatry, and Medicine at McGill, Montreal, QC, Canada
| | | | | | | | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, University College London Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at University College London, 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
| |
Collapse
|
28
|
Xiong SY, Zhao YL, Fu Y, Li QY, Hao Q, Zhang DD, Liu M, Yin S, Wang LY, Wang YC, Qiu SD, Zhang ZQ, Tan L. Associations between platelet indices and cerebrospinal fluid biomarkers of Alzheimer's disease pathology in cognitively intact adults: the CABLE study. Alzheimers Res Ther 2025; 17:124. [PMID: 40450373 DOI: 10.1186/s13195-025-01755-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 05/05/2025] [Indexed: 06/03/2025]
Abstract
INTRODUCTION Although previous studies have shown that specific platelet indices had correlations with cognitive impairment, the associations between platelet indices and cerebrospinal fluid (CSF) biomarkers of Alzheimer's disease (AD) pathology remain unclear. METHODS Our study included 1,047 cognitively normal participants from the Chinese Alzheimer's Biomarker and LifestylE (CABLE) study. The total participants had an average age of 58.33 years, a female proportion of 41.5% and average educational attainment of 9.58 years. Multiple linear regression models were used to analyze the associations of five platelet indices (plateletcrit [PCT], platelet count [PLT], mean platelet volume [MPV], platelet distribution width [PDW], and platelet large cell ratio [PLCR]) with CSF AD biomarkers after adjusting for age, gender, education and APOE ε4 allele status. Furthermore, the interactive, stratified and sensitivity analyses were further conducted to verify their relationships. RESULTS In the total participants, we found higher PCT levels were significantly correlated with higher CSF P-tau/Aβ42 (β = 0.102, P = 0.008) and T-tau/Aβ42 (β = 0.102, P = 0.008), as well as lower CSF Aβ42 (β = -0.089, P = 0.018) and Aβ42/Aβ40 (β = -0.093, P = 0.018). Moreover, other four platelet indices (PLT, MPV, PDW, PLCR) demonstrated moderate correlations with CSF AD biomarkers. The interaction analyses revealed that age affected the correlations between PCT and PLT with CSF Aβ42. Importantly, the associations between PCT and the aforementioned CSF AD biomarkers became more significant in the late-life group, but turned non-significant in the mid-life group. Besides, sensitivity analyses confirmed the robustness of our findings. CONCLUSIONS Our study provided preliminary evidence suggesting potential associations between platelet indices (especially PCT) and CSF AD biomarkers in cognitively intact adults. Nonetheless, more studies are needed to further validate these findings.
Collapse
Affiliation(s)
- Shi-Yin Xiong
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Yong-Li Zhao
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Yan Fu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Qiong-Yao Li
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Quan Hao
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Dan-Dan Zhang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Min Liu
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China
| | - Shan Yin
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Lan-Yang Wang
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Qingdao, China
| | - Yong-Chang Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China
| | - Shu-Dong Qiu
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Qingdao, China
| | - Zi-Qi Zhang
- School of Clinical Medicine, Weifang Medical University, Weifang, 261000, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, No.5 Donghai Middle Road, Qingdao, China.
| |
Collapse
|
29
|
Siebrand CJ, Bergo NJ, Lee S, Andersen JK, Walton CC. Chimeric antigen receptors discriminate between tau and distinct amyloid-beta species. J Transl Med 2025; 23:605. [PMID: 40448101 DOI: 10.1186/s12967-025-06572-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Accepted: 04/30/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND The lack of a definitive cure for Alzheimer's disease (AD) is fueling the search for innovative therapeutic strategies. Having revolutionized cancer immunotherapy, immune cell engineering with chimeric antigen receptors (CAR) is being explored to target AD. Whether CARs can recognize distinct amyloid-β (Aβ) species and tau neurofibrillary tangles (NFTs)-hallmark pathologies of AD-remains unclear. METHODS To investigate this, we engineered a series of CARs using single-chain fragment variable (scFv) derived from the variable light and heavy chains of antibodies tested in AD clinical trials. These included E2814 (E2814-CAR), targeting tau; Lecanemab (Lec-CAR) and Aducanumab (Adu-CAR), targeting Aβ; and Donanemab (Don-CAR) and Remternetug (Rem-CAR), targeting the truncated pyroglutamated Aβ species Aβp3-42. To evaluate CAR function, we utilized the murine DO11.10 CD4⁺ T-cell hybridoma line as a scalable and reproducible platform. CAR activation was assessed in response to tau preformed fibrils (PFFs), Aβ1-42 oligomer-enriched aggregates, and Aβp3-42 aggregates, using flow cytometry for CD69 expression and ELISA for IL-2 secretion. To validate this platform, we tested Adu-CAR in primary mouse CD4⁺ T cells treated with Aβ1-42 aggregates and assessed activation via flow cytometry for CD69 and CD25 expression. RESULTS DO11.10 cells expressing E2814-CAR-but not Lec-CAR-responded to tau PFFs. In contrast, cells expressing Adu-CAR, and to a lesser extent Lec-CAR-but not E2814-CAR-responded to Aβ1-42 aggregates. For Aβp3-42 aggregates, Rem-CAR elicited the strongest response, followed by Adu-CAR, while E2814-CAR and Don-CAR showed no activation. The activation of Adu-CAR by Aβ1-42 aggregates was recapitulated in primary CD4⁺ T cells, as measured by CD69 expression. CONCLUSIONS Our findings demonstrate that CARs can detect and discriminate between tau PFFs, Aβ1-42, and Aβp3-42 aggregates. This highlights the potential of repurposing AD antibodies for CAR-based therapies to selectively target tau NFTs and distinct forms of Aβ senile plaques.
Collapse
Affiliation(s)
- Cynthia J Siebrand
- Buck Institute for Research On Aging, 8001 Redwood Blvd., Novato, CA, 94945, USA
- USC Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Ave, Los Angeles, CA, 90191, USA
| | - Nicholas J Bergo
- Buck Institute for Research On Aging, 8001 Redwood Blvd., Novato, CA, 94945, USA
| | - Suckwon Lee
- Buck Institute for Research On Aging, 8001 Redwood Blvd., Novato, CA, 94945, USA
| | - Julie K Andersen
- Buck Institute for Research On Aging, 8001 Redwood Blvd., Novato, CA, 94945, USA.
| | - Chaska C Walton
- Buck Institute for Research On Aging, 8001 Redwood Blvd., Novato, CA, 94945, USA.
| |
Collapse
|
30
|
Fan KC, Lin CC, Chiu YL, Koh SH, Liu YC, Chuang YF. Compositional and functional gut microbiota alterations in mild cognitive impairment: links to Alzheimer's disease pathology. Alzheimers Res Ther 2025; 17:122. [PMID: 40448221 PMCID: PMC12123878 DOI: 10.1186/s13195-025-01769-9] [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: 03/18/2025] [Accepted: 05/19/2025] [Indexed: 06/02/2025]
Abstract
BACKGROUND Emerging evidence highlights the bidirectional communication between the gut microbiota and the brain, suggesting a potential role for gut dysbiosis in Alzheimer's disease (AD) pathology and cognitive decline. Existing literature on gut microbiota lacks species-level insights. This study investigates gut microbiota alterations in mild cognitive impairment (MCI), focusing on their association with comprehensive AD biomarkers, including amyloid burden, tau pathology, neurodegeneration, and cognitive performance. METHODS We analyzed fecal samples from 119 individuals with MCI and 320 cognitively normal controls enrolled in the Taiwan Precision Medicine Initiative on Cognitive Impairment and Dementia cohort. Shotgun metagenomic sequencing was conducted with taxonomic profiling using MetaPhlAn4. Amyloid burden and plasma pTau181 were quantified via PET imaging and Simoa assays, respectively, while APOE genotyping was performed using TaqMan assays. Microbial diversity, differential abundance analysis, and correlation mapping with neuropsychological and neuroimaging measures were conducted to identify gut microbiota species signatures associated with MCI and AD biomarkers. RESULTS We identified 59 key microbial species linked to MCI and AD biomarkers. Notably, species within the same genera, such as Bacteroides and Ruminococcus, showed opposing effects, while Akkermansia muciniphila correlated with reduced amyloid burden, suggesting a protective role. Functional profiling revealed microbial pathways contributing to energy metabolism and neuroinflammation, mediating the relationship between gut microbes and brain health. Co-occurrence network analyses demonstrated complex microbial interactions, indicating that the collective influence of gut microbiota on neurodegeneration. CONCLUSIONS Our findings challenge genus-level microbiome analyses, revealing species-specific modulators of AD pathology. This study highlights gut microbial activity as a potential therapeutic target to mitigate cognitive decline and neurodegeneration.
Collapse
Affiliation(s)
- Kang-Chen Fan
- School of Medicine, National Yang Ming Chiao Tung University, 155 Sec. 2, Linong St., Beitou Dist, Taipei City, 112304, Taiwan
| | - Chen-Ching Lin
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, 155 Sec. 2, Linong St., Beitou Dist, Taipei City, 112304, Taiwan
| | - Yen-Ling Chiu
- Department of Medical Research, Far Eastern Memorial Hospital, 21 Sec. 2, Nanya S. Rd., Banqiao Dist, New Taipei City, 220216, Taiwan
- Graduate Program in Biomedical Informatics, Graduate Institute of Medicine, Yuan Ze University, 135 Yuan-Tung Rd., Zhongli Dist, Taoyuan City, 32003, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University, 1 Sec. 4, Roosevelt Rd., Da'an Dist, Taipei City, 106319, Taiwan
| | - Seong-Ho Koh
- Department of Neurology, College of Medicine, Hanyang University Guri Hospital, 153 Kyougchun-ro, Guri-si, 11923, Gyeonggi-do, Republic of Korea
- Department of Translational Medicine, Graduate School of Biomedical Science & Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea
| | - Yi-Chien Liu
- Department of Neurology, Cardinal Tien Hospital, 362 Zhongzheng Rd., Xindian Dist, New Taipei City, 23148, Taiwan
| | - Yi-Fang Chuang
- School of Medicine, National Yang Ming Chiao Tung University, 155 Sec. 2, Linong St., Beitou Dist, Taipei City, 112304, Taiwan.
- Institute of Public Health, National Yang Ming Chiao Tung University, 155 Sec. 2, Linong St., Beitou Dist, Taipei City, 112304, Taiwan.
- Department of Psychiatry, Far Eastern Memorial Hospital, 21 Sec. 2, Nanya S. Rd., Banqiao Dist, New Taipei City, 220216, Taiwan.
| |
Collapse
|
31
|
Li Y, Zhou G, Peng J, Liu L, Zhang F, Iturria-Medina Y, Yao D, Biswal BB, Wang P. White matter dysfunction in Alzheimer's disease is associated with disease-related transcriptomic signatures. Commun Biol 2025; 8:820. [PMID: 40437109 PMCID: PMC12120127 DOI: 10.1038/s42003-025-08177-7] [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: 07/01/2024] [Accepted: 05/06/2025] [Indexed: 06/01/2025] Open
Abstract
While anatomical white matter (WM) alterations in Alzheimer's disease (AD) are well-established, functional WM dysregulation remains rarely investigated. The current study examines WM functional connectivity and network properties alterations in AD and mild cognitive impairment (MCI) and further describes their spatially correlated genes. AD and MCI shared decreased functional connectivity, clustering coefficient, and local efficiency within WM regions involved in impaired sensory-motor, visual-spatial, language, or memory functions. AD-specific dysfunction (i.e., AD vs. MCI and cognitively unimpaired participants) was predominantly located in WM, including anterior and posterior limb of internal capsule, corona radiata, and left tapetum. This WM dysfunction spatially correlates with specific genes, which are enriched in multiple biological processes related to synaptic function and development, and are mostly active in neurons and astrocytes. These findings may contribute to understanding molecular, cellular, and functional signatures associated with WM damage in AD.
Collapse
Affiliation(s)
- Yilu Li
- MOE Key Laboratory for Neuroinformation, The Clinical Hospital of Chengdu Brain Science Institute, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Guanyu Zhou
- MOE Key Laboratory for Neuroinformation, The Clinical Hospital of Chengdu Brain Science Institute, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Jinzhong Peng
- MOE Key Laboratory for Neuroinformation, The Clinical Hospital of Chengdu Brain Science Institute, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Lin Liu
- MOE Key Laboratory for Neuroinformation, The Clinical Hospital of Chengdu Brain Science Institute, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Fanyu Zhang
- MOE Key Laboratory for Neuroinformation, The Clinical Hospital of Chengdu Brain Science Institute, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Yasser Iturria-Medina
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, QC, Canada
| | - Dezhong Yao
- MOE Key Laboratory for Neuroinformation, The Clinical Hospital of Chengdu Brain Science Institute, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Bharat B Biswal
- MOE Key Laboratory for Neuroinformation, The Clinical Hospital of Chengdu Brain Science Institute, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China.
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA.
| | - Pan Wang
- MOE Key Laboratory for Neuroinformation, The Clinical Hospital of Chengdu Brain Science Institute, Center for Information in Medicine, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
32
|
Athalye C, Bahena A, Khandelwal P, Emrani S, Trotman W, Levorse LM, Khodakarami Z, Ohm DT, Teunissen-Bermeo E, Capp N, Sadaghiani S, Arezoumandan S, Lim SA, Prabhakaran K, Ittyerah R, Robinson JL, Schuck T, Lee EB, Tisdall MD, Das SR, Wolk DA, Irwin DJ, Yushkevich PA. Operationalizing postmortem pathology-MRI association studies in Alzheimer's disease and related disorders with MRI-guided histology sampling. Acta Neuropathol Commun 2025; 13:120. [PMID: 40437594 PMCID: PMC12121285 DOI: 10.1186/s40478-025-02030-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Accepted: 05/05/2025] [Indexed: 06/01/2025] Open
Abstract
Postmortem neuropathological examination, while the gold standard for diagnosing neurodegenerative diseases, often relies on limited regional sampling that may miss critical areas affected by Alzheimer's disease and related disorders. Ultra-high resolution postmortem MRI can help identify regions that fall outside the diagnostic sampling criteria for additional histopathologic evaluation. However, there are no standardized guidelines for integrating histology and MRI in a traditional brain bank. We developed a comprehensive protocol for whole hemisphere postmortem 7T MRI-guided histopathological sampling with whole-slide digital imaging and histopathological analysis, providing a reliable pipeline for high-volume brain banking in heterogeneous brain tissue. Our method uses patient-specific 3D printed molds built from postmortem MRI, allowing standardized tissue processing with a permanent spatial reference frame. To facilitate pathology-MRI association studies, we created a semi-automated MRI to histology registration pipeline and developed a quantitative pathology scoring system using weakly supervised deep learning. We validated this protocol on a cohort of 29 brains with diagnosis on the AD spectrum that revealed correlations between cortical thickness and phosphorylated tau accumulation. This pipeline has broad applicability across neuropathological research and brain banking, facilitating large-scale studies that integrate histology with neuroimaging. The innovations presented here provide a scalable and reproducible approach to studying postmortem brain pathology, with implications for advancing diagnostic and therapeutic strategies for Alzheimer's disease and related disorders.
Collapse
Affiliation(s)
- Chinmayee Athalye
- Department of Bioengineering, University of Pennsylvania, Philadelphia, USA.
| | - Alejandra Bahena
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | - Pulkit Khandelwal
- Department of Bioengineering, University of Pennsylvania, Philadelphia, USA
| | - Sheina Emrani
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | - Winifred Trotman
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | - Lisa M Levorse
- Department of Radiology, University of Pennsylvania, Philadelphia, USA
| | - Zahra Khodakarami
- Department of Bioengineering, University of Pennsylvania, Philadelphia, USA
| | - Daniel T Ohm
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | | | - Noah Capp
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | | | | | - Sydney A Lim
- Department of Radiology, University of Pennsylvania, Philadelphia, USA
| | | | - Ranjit Ittyerah
- Department of Radiology, University of Pennsylvania, Philadelphia, USA
| | - John L Robinson
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, USA
| | - Theresa Schuck
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, USA
| | - Edward B Lee
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, USA
| | - M Dylan Tisdall
- Department of Radiology, University of Pennsylvania, Philadelphia, USA
| | - Sandhitsu R Das
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | - David A Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | - David J Irwin
- Department of Neurology, University of Pennsylvania, Philadelphia, USA
| | - Paul A Yushkevich
- Department of Radiology, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
33
|
Sun J, Geng W, Wang Y, Li H, Tan R, Tu Y. An innovative electrochemiluminescent immunosensor using dual amplified signals from AuNPs@CoSn(OH) 6 for the detection of the AD biomarker: amyloid beta 1-40. Analyst 2025; 150:2259-2269. [PMID: 40326625 DOI: 10.1039/d5an00048c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Alzheimer's disease (AD) is a degenerative condition of the nervous system that causes severe damage to patients' daily activities and quality of life. Amyloid beta 1-40 protein (Aβ40), which is involved in the formation of cerebral plaques, is one of the crucial biomarkers related to AD. Herein, a novel and highly sensitive immunosensor for the detection of Aβ40 is developed. Using a reinforced indium tin oxide-coated glass with a nanocomposite of gold nanoparticle-enhanced CoSn(OH)6 (AuNPs@CoSn(OH)6) to trigger the electrochemiluminescence (ECL) of luminol as the sensing signal, the immunosensor is fabricated by immobilizing the Aβ40 antibody onto it. By integrating the high immune specificity, excellent conductivity and catalytic activity of the nanocomposite, the resultant immunosensor can be successfully employed to detect the target in real samples. The formation of the immune complex leads to increased steric hindrance and electron transfer resistance, which in turn causes a declined ECL output when the target Aβ40 binds to the antibody on the sensor surface. Under optimized conditions, the developed ECL immunosensor exhibits a linear response for Aβ40 ranging from 1 to 800 pg mL-1 and a low detection limit of 0.47 pg mL-1. Experimentally, it is demonstrated to be highly sensitive, specific, reproducible and stable. This work extends the application of the perovskite CoSn(OH)6 and AuNPs in the field of ECL immunosensing and provides a novel strategy for clinical research on Alzheimer's disease.
Collapse
Affiliation(s)
- Jiaojing Sun
- College of Chemistry, Chemical Engineering and Material Science, Soochow University, Suzhou, 215123, P. R. China.
| | - Wenqing Geng
- First Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China
| | - Yueju Wang
- First Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China
| | - Huiling Li
- First Affiliated Hospital of Soochow University, Suzhou, 215006, P. R. China
- Nursing School, Suzhou Medical College of Soochow University, Suzhou, 215006, P. R. China
| | - Rong Tan
- College of Chemistry, Chemical Engineering and Material Science, Soochow University, Suzhou, 215123, P. R. China.
- School of Material Engineering, Changshu Institute of Technology, Suzhou, 215500, P. R. China.
| | - Yifeng Tu
- College of Chemistry, Chemical Engineering and Material Science, Soochow University, Suzhou, 215123, P. R. China.
| |
Collapse
|
34
|
Bonham LW, Sirkis DW, Pang APS, Sugrue LP, Santamaría-García H, Ibáñez AM, Miller BL, Yokoyama JS, Corley MJ, for the Alzheimer’s Disease Neuroimaging Initiative. DNA methylation age from peripheral blood predicts progression to Alzheimer's disease, white matter disease burden, and cortical atrophy. NPJ DEMENTIA 2025; 1:7. [PMID: 40443531 PMCID: PMC12116384 DOI: 10.1038/s44400-025-00007-1] [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] [Received: 10/16/2024] [Accepted: 03/21/2025] [Indexed: 06/02/2025]
Abstract
Cross-sectional studies suggest a limited relationship between accelerated epigenetic aging derived from epigenetic clocks, and Alzheimer's disease (AD) pathophysiology or risk. However, most prior analyses have not utilized longitudinal analyses or whole-brain neuroimaging biomarkers of AD. Herein, we employed longitudinal modeling and structural neuroimaging analyses to test the hypothesis that accelerated epigenetic aging would predict AD progression. Using survival analyses, we found that two second-generation epigenetic clocks, DNAmPhenoAge and DNAmGrimAge, predicted progression from cognitively normal aging to mild cognitive impairment or AD and worse longitudinal cognitive outcomes. Epigenetic age was also strongly associated with cortical thinning in AD-relevant regions and white matter disease burden. Thus, in contrast to earlier work suggesting limited applicability of blood-based epigenetic clocks in AD, our novel analytic framework suggests that second-generation epigenetic clocks have broad utility and may represent promising predictors of AD risk and pathophysiology.
Collapse
Affiliation(s)
- Luke W. Bonham
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA USA
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA USA
| | - Daniel W. Sirkis
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA USA
| | - Alina P. S. Pang
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY USA
| | - Leo P. Sugrue
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA USA
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA USA
| | - Hernando Santamaría-García
- Pontificia Universidad Javeriana, Physiology and Psychiatry Departments, Bogotá, Colombia
- Centro de Memoria y Cognición Intellectus, Hospital Universitario San Ignacio, Bogotá, Colombia
| | - Agustín M. Ibáñez
- Center for Social and Cognitive Neuroscience, School of Psychology, Universidad Adolfo Ibáñez, Santiago de Chile, Chile
- Universidad de San Andrés, Buenos Aires, Argentina
- National Scientific and Technical Research Council, Buenos Aires, Argentina
| | - Bruce L. Miller
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA USA
- Global Brain Health Institute, University of California San Francisco, San Francisco, CA USA
| | - Jennifer S. Yokoyama
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA USA
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA USA
- Global Brain Health Institute, University of California San Francisco, San Francisco, CA USA
| | - Michael J. Corley
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY USA
- Division of Geriatrics, Gerontology & Palliative Care, Department of Medicine, University of California, San Diego, La Jolla, CA USA
| | - for the Alzheimer’s Disease Neuroimaging Initiative
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA USA
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA USA
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY USA
- Department of Psychiatry, University of California, San Francisco, San Francisco, CA USA
- Pontificia Universidad Javeriana, Physiology and Psychiatry Departments, Bogotá, Colombia
- Centro de Memoria y Cognición Intellectus, Hospital Universitario San Ignacio, Bogotá, Colombia
- Center for Social and Cognitive Neuroscience, School of Psychology, Universidad Adolfo Ibáñez, Santiago de Chile, Chile
- Universidad de San Andrés, Buenos Aires, Argentina
- National Scientific and Technical Research Council, Buenos Aires, Argentina
- Global Brain Health Institute, University of California San Francisco, San Francisco, CA USA
- Division of Geriatrics, Gerontology & Palliative Care, Department of Medicine, University of California, San Diego, La Jolla, CA USA
| |
Collapse
|
35
|
Xiang J, Qian Z, Xi Y, Wei Y, Wang G, Liu X, Wang ZH, Zhang Z, Wu S, Ye K. Immunotherapy against tau fragment diminishes AD pathology, improving synaptic function and cognition. Mol Neurodegener 2025; 20:60. [PMID: 40426267 PMCID: PMC12117789 DOI: 10.1186/s13024-025-00854-9] [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: 12/15/2024] [Accepted: 05/20/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Asparagine endopeptidase (AEP) is implicated in the pathogenesis of Alzheimer's disease (AD) by cleaving Tau at residue N368, accelerating its hyperphosphorylation and aggregation. The Tau N368/t-Tau ratio in cerebrospinal fluid (CSF) serves as a superior biomarker compared to established biomarkers (p-Tau 181/217) for correlating with tau pathology and synaptic dysfunction in patients with AD, highlighting its diagnostic and therapeutic potential. METHODS We evaluated the therapeutic efficacy of a Tau N368-specific antibody in two mouse models: Tau P301S (tauopathy) and 3xTg (AD with Aβ/tau pathology). We conducted chronic intraperitoneal administration of the antibody to evaluate its effects on tau aggregation, synaptic integrity, and cognitive function. Neuropathological changes, synaptic plasticity (through electrophysiology), and behavioral outcomes were analyzed alongside Aβ pathology and neuroinflammation in 3xTg mice. RESULTS Treatment with the anti-Tau N368 antibody significantly diminished neurofibrillary tangles (NFTs) formed of hyperphosphorylated/truncated Tau in both models. Clearance of Tau restored BDNF/TrkB neurotrophic signaling, improved synaptic plasticity, and alleviated cognitive deficits. In 3xTg mice, this treatment also reduced Aβ deposition and neuroinflammation, resulting in enhanced learning and memory. Notably, the antibody's effectiveness in alleviating both tau and Aβ pathologies indicates a potential interaction between these pathways. CONCLUSIONS Targeting Tau N368 through immunotherapy alleviates tau-driven neurodegeneration, restores synaptic function, and improves accompanying Aβ pathology in AD models. Our results confirmed that Tau N368 is an exceptional biomarker and a promising therapeutic target, disrupting AD progression by addressing tau aggregation and its downstream effects.
Collapse
Affiliation(s)
- Jie Xiang
- Department of Neurobiology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Zhengjiang Qian
- Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, Guangdong, 518055, China
| | - Ye Xi
- Department of Neurobiology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Yanuo Wei
- Department of Neurobiology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Guangxing Wang
- Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Xia Liu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Zhi-Hao Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Shengxi Wu
- Department of Neurobiology, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China.
| | - Keqiang Ye
- Department of Biology, Faculty of Life & Health Sciences, Shenzhen University of Advanced Technology (SUAT), Shenzhen, China.
| |
Collapse
|
36
|
van den Berg RL, van der Landen SM, Keijzer MJ, van Gils AM, van Dam M, Ziesemer KA, Jutten RJ, Harrison JE, de Boer C, van der Flier WM, Sikkes SA. Smartphone- and Tablet-Based Tools to Assess Cognition in Individuals With Preclinical Alzheimer Disease and Mild Cognitive Impairment: Scoping Review. J Med Internet Res 2025; 27:e65297. [PMID: 40424609 DOI: 10.2196/65297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 01/24/2025] [Accepted: 02/24/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Assessment of cognitive decline in the earliest stages of Alzheimer disease (AD) is important but challenging. AD is a neurodegenerative disease characterized by gradual cognitive decline. Disease stages range from preclinical AD, in which individuals are cognitively unimpaired, to mild cognitive impairment (MCI) and dementia. Digital technologies promise to enable detection of early, subtle cognitive changes. Although the field of digital cognitive biomarkers is rapidly evolving, a comprehensive overview of the reporting of psychometric properties (ie, validity, reliability, responsiveness, and clinical meaningfulness) is missing. Insight into the extent to which these properties are evaluated is needed to identify the validation steps toward implementation. OBJECTIVE This scoping review aimed to identify the reporting on quality characteristics of smartphone- and tablet-based cognitive tools with potential for remote administration in individuals with preclinical AD or MCI. We focused on both psychometric properties and practical tool characteristics. METHODS This scoping review was conducted following the PRISMA-ScR (Preferred Reporting Items for Systematic Reviews and Meta-Analyses extension for Scoping Reviews) guidelines. In total, 4 databases (PubMed, Embase, Web of Science, and PsycINFO) were systematically searched from January 1, 2008, to January 5, 2023. Studies were included that assessed the psychometric properties of cognitive smartphone- or tablet-based tools with potential for remote administration in individuals with preclinical AD or MCI. In total, 2 reviewers independently screened titles and abstracts in ASReview, a screening tool that combines manual and automatic screening using an active learning algorithm. Thereafter, we manually screened full texts in the web application Rayyan. For each included study, 2 reviewers independently explored the reported information on practical and psychometric properties. For each psychometric property, examples were provided narratively. RESULTS In total, 11,300 deduplicated studies were identified in the search. After screening, 50 studies describing 37 different digital tools were included in this review. Average administration time was 13.8 (SD 10.1; range 1-32) minutes, but for 38% (14/37) of the tools, this was not described. Most tools (31/37, 84%) were examined in 1 language. The investigated populations were mainly individuals with MCI (34/37, 92%), and fewer tools were examined in individuals with preclinical AD (8/37, 22%). For almost all tools (36/37, 97%), construct validity was assessed through evaluation of clinical or biological associations or relevant group differences. For a small number of tools, information on structural validity (3/37, 8%), test-retest reliability (12/37, 32%), responsiveness (6/37, 16%), or clinical meaningfulness (0%) was reported. CONCLUSIONS Numerous smartphone- and tablet-based tools to assess cognition in early AD are being developed, whereas studies concerning their psychometric properties are limited. Often, initial validation steps have been taken, yet further validation and careful selection of psychometrically valid outcome scores are required to demonstrate clinical usefulness with regard to the context of use, which is essential for implementation.
Collapse
Affiliation(s)
- Rosanne L van den Berg
- Alzheimer Center Amsterdam, Neurology, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Neurodegeneration, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Clinical, Neuro and Developmental Psychology, Faculty of Movement and Behavioral Sciences, VU University, Amsterdam, The Netherlands
| | - Sophie M van der Landen
- Alzheimer Center Amsterdam, Neurology, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Neurodegeneration, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Matthijs J Keijzer
- Alzheimer Center Amsterdam, Neurology, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Neurodegeneration, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Neurocast BV, Amsterdam, The Netherlands
| | - Aniek M van Gils
- Alzheimer Center Amsterdam, Neurology, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Neurodegeneration, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Maureen van Dam
- Alzheimer Center Amsterdam, Neurology, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Neurodegeneration, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | | | - Roos J Jutten
- Alzheimer Center Amsterdam, Neurology, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Neurodegeneration, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - John E Harrison
- Alzheimer Center Amsterdam, Neurology, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Neurodegeneration, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Metis Cognition Ltd., Kilmington Common, United Kingdom
- Department of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Casper de Boer
- Alzheimer Center Amsterdam, Neurology, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Neurodegeneration, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Neurodegeneration, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Epidemiology and Biostatistics, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | - Sietske Am Sikkes
- Alzheimer Center Amsterdam, Neurology, Amsterdam University Medical Center, Amsterdam, The Netherlands
- Neurodegeneration, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Clinical, Neuro and Developmental Psychology, Faculty of Movement and Behavioral Sciences, VU University, Amsterdam, The Netherlands
| |
Collapse
|
37
|
Aksnes M. Sex Differences in Biofluid Biomarkers for Alzheimer's Disease. NEURODEGENER DIS 2025:1-11. [PMID: 40418910 DOI: 10.1159/000545717] [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: 09/30/2024] [Accepted: 04/01/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD), the most common cause of dementia, affects twice as many women as men. Moreover, sex is increasingly recognised as an important factor for AD, influencing symptom presentation, progression, disease biology, and treatment responses. In parallel, AD biomarkers are becoming more accessible with the discovery of specific and accurate blood-based biomarkers and their incorporation in AD diagnostic frameworks. This narrative review aimed to summarise sex differences in the concentration and interpretation of biofluid biomarkers for AD. SUMMARY Biological sex may influence both the concentration and interpretation of biofluid biomarkers for AD pathology such as amyloid-β aggregation, tau neurofibrillary tangles, neurodegeneration, or neuroinflammation. While some biofluid biomarkers display consistent sex differences in absolute levels, most biomarker levels have not been found to differ consistently by sex. Nonetheless, even biomarkers that do not differ in absolute levels display sex-specific associations with clinically relevant variables such as brain atrophy, cognitive impairment, and disease progression. KEY MESSAGE Sex may influence the interpretation of AD biomarkers depending on their context of use, and more research is required to develop sex-specific guidelines. Future research should aim to study sex differences and sex-specific associations with variables of interest, as well as the underlying factors driving sex differences in AD.
Collapse
Affiliation(s)
- Mari Aksnes
- Department of Geriatric Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
38
|
Choudry M, Gould M, Ganti L. Treatment of agitation in dementia - a systematic review. Int J Emerg Med 2025; 18:101. [PMID: 40414837 DOI: 10.1186/s12245-025-00902-7] [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: 02/09/2025] [Accepted: 05/15/2025] [Indexed: 05/27/2025] Open
Abstract
Dementia, including Alzheimer's disease (AD), affects millions worldwide, leading to cognitive decline, memory loss, and behavioral disturbances. Agitation, a common symptom, poses significant challenges for patients and caregivers. This systematic review evaluates pharmacological and non-pharmacological interventions for managing agitation in dementia. Nine clinical studies were analyzed, encompassing medications like brexpiprazole and non-pharmacological approaches such as music therapy and digital care programs. Pharmacological treatments showed mixed efficacy and safety profiles, with brexpiprazole demonstrating dose-dependent benefits and mirtazapine associating with higher mortality rates. Non-pharmacological interventions exhibited promising results in reducing agitation without adverse effects. The review underscores the importance of personalized care strategies tailored to individual patient needs and preferences to enhance treatment efficacy and quality of life, highlighting the shift toward holistic, individualized care.
Collapse
Affiliation(s)
| | - Murdoc Gould
- Birkbeck, University of London, Malet St, London, WC1E 7HX, UK
| | - Latha Ganti
- Orlando College of Osteopathic Medicine, Winter Garden, FL, 34787, USA.
- Warren Alpert Medical School of Brown University, 222 Richmond Street, Providence, RI, 02903, USA.
| |
Collapse
|
39
|
Delhaye E, Besson G, Bahri MA, Bastin C. Object fine-grained discrimination as a sensitive cognitive marker of transentorhinal integrity. Commun Biol 2025; 8:800. [PMID: 40415135 DOI: 10.1038/s42003-025-08201-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 05/08/2025] [Indexed: 05/27/2025] Open
Abstract
The transentorhinal cortex (tErC) is one of the first regions affected by Alzheimer's disease (AD), often showing changes before clinical symptoms appear. Understanding its role in cognition is key to detecting early cognitive impairments in AD. This study tested the hypothesis that the tErC supports fine-grained representations of unique individual objects, sensitively to the granularity of the demanded discrimination, influencing both perceptual and mnemonic functions. We examined the tErC's role in object versus scene discrimination, using objective (based on a pretrained convolutional neural network, CNN) and subjective (human-rated) measures of visual similarity. Our results show that the structural integrity of the tErC is specifically related to the sensitivity to visual similarity for objects, but not for scenes. Importantly, this relationship depends on how visual similarity is measured: it appears only when using CNN visual similarity measures in perceptual discrimination, and solely when using subjective similarity ratings in mnemonic discrimination. Furthermore, in mnemonic discrimination, object sensitivity to visual similarity was specifically associated with the integrity of tErC-BA36 connectivity, only when similarity was computed from subjective ratings. Altogether, these findings suggest that discrimination sensitivity to object visual similarity may represent a specific marker of tErC integrity after accounting for the type of similarity measured.
Collapse
Affiliation(s)
- Emma Delhaye
- GIGA Research, CRC Human Imaging, University of Liège, Liège, Belgium.
- PsyNCog Research Unit, Faculty of Psychology, University of Liège, Liège, Belgium.
- CICPSI, Faculty of Psychology, University of Lisbon, Lisbon, Portugal.
| | - Gabriel Besson
- Proaction Laboratory, Faculty of Psychology and Educational Sciences, University of Coimbra, Coimbra, Portugal
- CINEICC, Faculty of Psychology and Educational Sciences, University of Coimbra, Coimbra, Portugal
| | - Mohamed Ali Bahri
- GIGA Research, CRC Human Imaging, University of Liège, Liège, Belgium
| | - Christine Bastin
- GIGA Research, CRC Human Imaging, University of Liège, Liège, Belgium
- PsyNCog Research Unit, Faculty of Psychology, University of Liège, Liège, Belgium
| |
Collapse
|
40
|
Frederiksen KS, Boada M, Dubois B, Engelborghs S, Frisoni GB, Georges J, Hort J, Jönsson L, Kramberger MG, Ousset PJ, Scarmeas N, Schmidt R, Schott JM, Spiro L, Waldemar G, Winblad B, Jessen F, Frölich L. Navigating the introduction of anti-amyloid therapy in Europe: a position statement by individual members of the EADC. Alzheimers Res Ther 2025; 17:116. [PMID: 40413561 PMCID: PMC12102950 DOI: 10.1186/s13195-025-01766-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 05/13/2025] [Indexed: 05/27/2025]
Abstract
INTRODUCTION Anti-amyloid antibodies for the treatment of Alzheimer´s disease (AD) are currently being evaluated for approval and reimbursement in Europe. An approval brings opportunities, but also challenges to health care systems across Europe. The objective of this position paper is to provide guidance from experts in the field in terms of navigating implementation. METHODS Members of the European Alzheimer's Disease Consortium and a representative of Alzheimer Europe convened to formulate recommendations covering key areas related to the possible implementation of anti-amyloid antibodies in AD through online discussions and 2 rounds of online voting with an 80% threshold for a position to be accepted. RESULTS In total, 24 recommendations were developed covering the research landscape and priorities within research in AD following a possible approval, potential impact on health care systems and diagnostic pathways, and communication to patients about anti-amyloid antibodies. Anti-amyloid antibodies are regarded as a substantial innovation with an important clinical impact. In addition, however, new compounds with other mechanisms of action and/or route of administration are also needed. Approval of new treatments will require changes to existing patient pathways and real-world data needs to be generated. CONCLUSION Comprehensive guidance is provided on the potential implementation of anti-amyloid antibody therapies in Europe following possible approval. Emphasis is placed on the necessity of regularly updating recommendations as new evidence emerges in the coming years.
Collapse
Affiliation(s)
- Kristian S Frederiksen
- Danish Dementia Research Centre, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Mercé Boada
- Ace Alzheimer Center Barcelona - Universitat Internacional de Catalunya, Barcelona, Spain
- Networking Research Center On Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Bruno Dubois
- Institute of Memory and Alzheimer's Disease (IM2 A), Department of Neurology, Assistance Publique-Hôpitaux de Paris (AP-HP), Sorbonne University, Pitié-Salpêtrière Hospital, Paris, France
- Frontlab, Paris Brain Institute (Institut du Cerveau, ICM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Sebastiaan Engelborghs
- Department of Neurology and Bru-BRAIN, NEUR Research Group, Center for Neurosciences (C4 N), Universitair Ziekenhuis Brusseland, VrijeUniversiteit Brussel (VUB) , Brussels, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Giovanni B Frisoni
- LANVIE - Laboratory of Neuroimaging of Aging, Memory Centerand, University Hospitals and University of Geneva , Geneva, Switzerland
| | | | - Jakub Hort
- Department of Neurology, Second Faculty of Medicine, Memory Clinic, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Linus Jönsson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Milica G Kramberger
- Department of Neurology, Faculty of Medicine, University Medical Center, University of Ljubljana, Ljubljana, Slovenia
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Pierre-Jean Ousset
- Toulouse Clinical Research Center, Toulouse University Hospital, Toulouse, France
| | - Nikolaos Scarmeas
- 1, Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School GR, Athens, Greece
- Department of Neurology, Taub Institute for Research in Alzheimer's Disease and the Aging Brain, The Gertrude H. Sergievsky Center, Columbia University, New York, NY, USA
| | - Reinhold Schmidt
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University Graz, Graz, Austria
| | - Jonathan M Schott
- Dementia Research Centre, UCL Queen Square Institute of Neurology, UCL, London, UK
| | - Luiza Spiro
- Carol Davila" University of Medicine and Pharmacy (www.umfcd.ro), Bucharest, Romania
- Faculty, Geneva College of Longevity Science (www.gcls.study), Geneva, Switzerland
- Faculty, Geneva School of Business Administration, University of Applied Sciences of Western Switzerland (www.hesge.ch), Geneva, Switzerland
- President "Ana ASLAN International" Foundation (www.anaaslanacademy.ro), Excellence Memory Center, Brain Health, and Longevity Medicine, Bucharest, Romania
| | - Gunhild Waldemar
- Danish Dementia Research Centre, Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bengt Winblad
- Aging and Inflammation Theme, Karolinska University Hospital, Stockholm, Sweden
- Division of Neurogeriatrics, Dept of Neurobiology, Care Sciences and Society, Theme Inflammation & Aging, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| | - Frank Jessen
- Department of Psychiatry, Medical Faculty, University of Cologne, Cologne, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Excellence Cluster On Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Lutz Frölich
- Department of Geriatric Psychiatry, Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| |
Collapse
|
41
|
Gaetani L, Bellomo G, Chiasserini D, De Rocker C, Goossens J, Paolini Paoletti F, Vanmechelen E, Parnetti L. Influence of co-pathology on CSF and plasma synaptic markers SNAP25 and VAMP2 in Alzheimer's disease and Parkinson's disease. Alzheimers Res Ther 2025; 17:115. [PMID: 40405270 PMCID: PMC12096548 DOI: 10.1186/s13195-025-01762-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 05/12/2025] [Indexed: 05/24/2025]
Abstract
BACKGROUND Synaptic dysfunction is a relevant feature of Alzheimer's disease (AD) and Parkinson's disease (PD) and can be quantified through the measurement of cerebrospinal fluid (CSF) synaptic markers, such as the presynaptic proteins synaptosomal-associated protein 25 kDa (SNAP25) and vesicle-associated membrane protein 2 (VAMP2). Plasma-based assays for synaptic markers are also emerging. In neurodegenerative diseases, synaptic dysfunction can be directly driven by proteinopathies such as amyloidosis (A), tauopathy (T), and α-synucleinopathy (S), which in turn can be detected via CSF biomarkers. This observational study aimed to: (i) evaluate the concordance of SNAP25 and VAMP2 in CSF and plasma; (ii) compare SNAP25 and VAMP2 concentrations in CSF and plasma across AD, PD, and control groups; (iii) examine the impact on synaptic markers concentration of CSF α-synuclein seed amplification assay (αS-SAA) positivity (S+) in AD, and (iv) of CSF amyloid/tau (A+/T+) positivity in PD. METHODS We included 80 AD patients (preclinical, mild cognitive impairment [MCI], and dementia stages), 47 PD patients, and 41 controls with other neurological diseases (OND) and known CSF A/T/S profiles. All AD and 5/47 PD patients were CSF A+/T+, while 26/80 AD and all PD patients were CSF S+. All OND had a non-A+/T + and a S - profile. SNAP25 and VAMP2 concentrations in CSF and plasma were measured using Simoa-based immunoassays. RESULTS CSF and plasma SNAP25 were positively correlated, but no correlation was observed for VAMP2 in these matrices. CSF and plasma SNAP25 and CSF VAMP2 were higher in AD compared to PD and OND. Synaptic markers were elevated in preclinical AD and remained stable across MCI-AD and AD dementia stages. AD patients with CSF αS-SAA positivity showed no significant difference in synaptic markers compared to those without CSF αS-SAA positivity, independent of clinical stage. In PD, A+/T + patients had higher CSF and plasma SNAP25 (132.3 ± 41.6; 1.9 ± 0.5 pg/mL) compared to non-A+/T + PD (105.4 ± 34.2; 1.3 ± 0.3 pg/mL) (p < 0.001 and p < 0.01, respectively). CONCLUSIONS SNAP25 reliably serves as a marker of synaptic injury when measured in both CSF and plasma, whereas VAMP2 demonstrates reliability exclusively in CSF. Both markers are primarily influenced by AD pathology and remain unaffected by α-synucleinopathy, suggesting their potential in detecting AD-related synaptic dysfunction.
Collapse
Affiliation(s)
- Lorenzo Gaetani
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giovanni Bellomo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Davide Chiasserini
- Section of Biochemistry, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Julie Goossens
- ADx NeuroSciences NV, Technologiepark Zwijnaarde 6, Ghent, 9052, Belgium
| | | | - Eugeen Vanmechelen
- ADx NeuroSciences NV, Technologiepark Zwijnaarde 6, Ghent, 9052, Belgium.
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
| |
Collapse
|
42
|
Meng W, Inampudi R, Zhang X, Xu J, Huang Y, Xie M, Bian J, Yin R. An Interpretable Population Graph Network to Identify Rapid Progression of Alzheimer's Disease Using UK Biobank. AMIA ... ANNUAL SYMPOSIUM PROCEEDINGS. AMIA SYMPOSIUM 2025; 2024:808-817. [PMID: 40417509 PMCID: PMC12099444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/27/2025]
Abstract
Alzheimer's disease (AD) manifests with varying progression rates across individuals, necessitating the understanding of their intricate patterns of cognition decline that could contribute to effective strategies for risk monitoring. In this study, we propose an innovative interpretable population graph network framework for identifying rapid progressors of AD by utilizing patient information from electronic health-related records in the UK Biobank. To achieve this, we first created a patient similarity graph, in which each AD patient is represented as a node; and an edge is established by patient clinical characteristics distance. We used graph neural networks (GNNs) to predict rapid progressors of AD and created a GNN Explainer with SHAP analysis for interpretability. The proposed model demonstrates superior predictive performance over the existing benchmark approaches. We also revealed several clinical features significantly associated with the prediction, which can be used to aid in effective interventions for the progression of AD patients.
Collapse
Affiliation(s)
- Weimin Meng
- Department of Health Outcomes and Biomedical Informatics, University of Florida, Gainesville, FL, USA
| | - Rohit Inampudi
- Department of Computer Science and Engineering, University of Florida, Gainesville, FL, USA
| | - Xiang Zhang
- Department of Computer Science, University of North Carolina at Charlotte, Charlotte, NC, US
| | - Jie Xu
- Department of Health Outcomes and Biomedical Informatics, University of Florida, Gainesville, FL, USA
| | - Yu Huang
- Department of Health Outcomes and Biomedical Informatics, University of Florida, Gainesville, FL, USA
| | - Mingyi Xie
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, USA
| | - Jiang Bian
- Department of Health Outcomes and Biomedical Informatics, University of Florida, Gainesville, FL, USA
| | - Rui Yin
- Department of Health Outcomes and Biomedical Informatics, University of Florida, Gainesville, FL, USA
| |
Collapse
|
43
|
Sugandhi VV, Gadhave DG, Ugale AR, Kulkarni N, Nangare SN, Patil HP, Rath S, Saxena R, Lavate A, Patel AT, Jadhav A, Paudel KR. Advances in Alzheimer's Therapy: Exploring Neuropathological Mechanisms to Revolutionize the Future Therapeutic Landscape. Ageing Res Rev 2025; 109:102775. [PMID: 40403980 DOI: 10.1016/j.arr.2025.102775] [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: 12/12/2024] [Revised: 05/07/2025] [Accepted: 05/16/2025] [Indexed: 05/24/2025]
Abstract
Alzheimer's disease (AD) is still an excessively complicated neurological disorder that impacts millions of individuals globally. The ideal defensive feature of the central nervous system (CNS) is the intimate junction of endothelial cells, which functions as a biological barrier to safely control molecular transport throughout the brain. The blood-brain barrier (BBB) comprises tightly locked astrocyte cell junctions on CNS blood capillaries. This biological barrier shields the brain from hazardous toxins by preventing the entry of polar medications, cells, and ions. However, it is very challenging to provide any treatment to the brain for neurodegenerative illnesses like Alzheimer's. Different causative mechanisms, such as amyloid-β (Aβ) plaques, tubulin-associated unit (Tau) tangles, and neuroinflammation, cause neuronal dysfunction, leading to dementia and memory loss in the subject. Several treatments are approved for AD therapy, whereas most only help treat related symptoms. Disappointingly, current remedies have not been able to control the progression of AD due to associated side effects. Specific pathogenic mechanisms are involved in the initiation and development of this disease. Therefore, the expected survival of a patient with AD is limited and is approximately ten years. Hence, the pathogenic mechanism behind AD progression must be understood to better comprehend and improve the overall survival rate. This review highlighted the recent insights into AD pathogenesis, molecular mechanisms, advancements in theragnostic techniques, the existing updates of clinical trials, and emerging innovations for AD medicinal development. That has helped researchers develop other strategies to address the shortcomings of traditional medications.
Collapse
Affiliation(s)
- Vrashabh V Sugandhi
- College of Pharmacy & Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Dnyandev G Gadhave
- College of Pharmacy & Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA; Department of Pharmaceutics, Dattakala Shikshan Sanstha's, Dattakala College of Pharmacy (Affiliated to Savitribai Phule Pune University), Swami Chincholi, Daund, Pune, Maharashtra 413130, India.
| | - Akanksha R Ugale
- College of Pharmacy & Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Nilesh Kulkarni
- Department of Pharmaceutics, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dhule, Maharashtra 425405, India
| | - Sopan N Nangare
- Department of Pharmaceutical Chemistry, H. R. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dhule, Maharashtra 425405, India
| | - Harshal P Patil
- Department of Pharmaceutics, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, Dhule, Maharashtra 425405, India
| | - Seepra Rath
- Center for Dermal Research, Rutgers, the State University of New Jersey, 145 Bevier Road, Piscataway 08854, USA
| | - Rahul Saxena
- Graduate Programs in Molecular Biosciences, Rutgers, the State University of New Jersey, Piscataway 08854, USA
| | - Amol Lavate
- Department of Pharmaceutics, Dattakala Shikshan Sanstha's, Dattakala College of Pharmacy (Affiliated to Savitribai Phule Pune University), Swami Chincholi, Daund, Pune, Maharashtra 413130, India
| | - Apeksha T Patel
- Department of Quality Assurance, Navinta III INC, Boca Raton, 33487, Florida, USA
| | - Ashish Jadhav
- Department of Pharmaceutics, Dattakala Shikshan Sanstha's, Dattakala College of Pharmacy (Affiliated to Savitribai Phule Pune University), Swami Chincholi, Daund, Pune, Maharashtra 413130, India
| | - Keshav Raj Paudel
- Centre for Inflammation, Faculty of Science, School of Life Science, Centenary Institute and University of Technology Sydney, Sydney 2007, Australia.
| |
Collapse
|
44
|
Heo G, Xu Y, Wang E, Ali M, Oh HSH, Moran-Losada P, Anastasi F, González Escalante A, Puerta R, Song S, Timsina J, Liu M, Western D, Gong K, Chen Y, Kohlfeld P, Flynn A, Thomas AG, Lowery J, Morris JC, Holtzman DM, Perlmutter JS, Schindler SE, Vilor-Tejedor N, Suárez-Calvet M, García-González P, Marquié M, Fernández MV, Boada M, Cano A, Ruiz A, Zhang B, Bennett DA, Benzinger T, Wyss-Coray T, Ibanez L, Sung YJ, Cruchaga C. Large-scale plasma proteomic profiling unveils diagnostic biomarkers and pathways for Alzheimer's disease. NATURE AGING 2025:10.1038/s43587-025-00872-8. [PMID: 40394224 DOI: 10.1038/s43587-025-00872-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 04/09/2025] [Indexed: 05/22/2025]
Abstract
Proteomic studies have been instrumental in identifying brain, cerebrospinal fluid and plasma proteins associated with Alzheimer's disease (AD). Here, we comprehensively examined 6,905 aptamers corresponding to 6,106 unique proteins in plasma in more than 3,300 well-characterized individuals to identify new proteins, pathways and predictive models for AD. We identified 416 proteins (294 new) associated with clinical AD status and validated the findings in two external datasets representing more than 7,000 samples. AD-related proteins reflected blood-brain barrier disruption and other processes implicated in AD, such as lipid dysregulation or immune responses. A machine learning model was used to identify a set of seven proteins that were highly predictive of both clinical AD (area under the curve (AUC) of >0.72) and biomarker-defined AD status (AUC of >0.88), which were replicated in multiple external cohorts and orthogonal platforms. These findings underscore the potential of using plasma proteins as biomarkers for the early detection and monitoring of AD and for guiding treatment decisions.
Collapse
Affiliation(s)
- Gyujin Heo
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, USA
| | - Ying Xu
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, USA
| | - Erming Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Muhammad Ali
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, USA
| | - Hamilton Se-Hwee Oh
- Graduate Program in Stem Cell and Regenerative Medicine, Stanford University, Stanford, CA, USA
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Patricia Moran-Losada
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - 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
| | - Armand González Escalante
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- Hospital del Mar Research Institute, Barcelona, Spain
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Raquel Puerta
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- PhD Program in Biotechnology, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Soomin Song
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, USA
| | - Jigyasha Timsina
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, USA
| | - Menghan Liu
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, USA
| | - Daniel Western
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, USA
| | - Katherine Gong
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, USA
| | - Yike Chen
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, USA
| | - Pat Kohlfeld
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, USA
| | - Allison Flynn
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, USA
| | - Alvin G Thomas
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, USA
| | - Joseph Lowery
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, USA
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - David M Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurologic Diseases, Washington University, St. Louis, MO, USA
- Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO, USA
| | - Joel S Perlmutter
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Suzanne E Schindler
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - 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 UMC, Nijmegen, Netherlands
| | - 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
| | - Pablo García-González
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, Madrid, Spain
| | - Marta Marquié
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, Madrid, Spain
| | - Maria Victoria Fernández
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, Madrid, Spain
| | - Mercè Boada
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, Madrid, Spain
| | - Amanda Cano
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, Madrid, Spain
| | - Agustín Ruiz
- Ace Alzheimer Center Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), National Institute of Health Carlos III, Madrid, Spain
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center, San Antonio, TX, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Tammie Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tony Wyss-Coray
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Laura Ibanez
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yun Ju Sung
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, USA
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA.
- NeuroGenomics and Informatics Center, Washington University, St. Louis, MO, USA.
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
- Hope Center for Neurologic Diseases, Washington University, St. Louis, MO, USA.
- Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO, USA.
| |
Collapse
|
45
|
Giannakis A, Konitsiotis S. Anti-Amyloid Agents: A Self-Fulfilling prophecy. J Clin Neurosci 2025; 137:111338. [PMID: 40393205 DOI: 10.1016/j.jocn.2025.111338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 05/15/2025] [Accepted: 05/15/2025] [Indexed: 05/22/2025]
Abstract
The introduction of anti-amyloid antibodies has ushered in a new era in the treatment of Alzheimer's disease (AD), coinciding with the revision of its diagnostic criteria, which now focus on the biological definition of AD, with amyloid beta at its core. However, despite being fully aligned with these criteria-and therefore with how we define the disease-amyloid-targeting therapies have not yielded the expected results. How can a treatment targeting the very core of the disease be ineffective? Perhaps because AD, as we have defined it, is not actually the disease that afflicts millions of patients worldwide. Patients with conditions related to AD, such as apolipoprotein ε4 allele (APOE4) homozygotes, patients receiving anticoagulant therapy for atrial fibrillation, and those with microhemorrhages, are excluded from treatment. Several other pathogenetic mechanisms continue to arise, including neuroinflammation, cerebrovascular disease, and metal ion dysregulation. At the same time, Alzheimer's pathology frequently coexists with other brain pathologies in AD patients, the roles and interactions of which remain largely unknown. Thus, AD should be redefined as a multifactorial neurodegenerative disorder, in which various processes contribute to amyloid accumulation or independently drive neurodegeneration.
Collapse
Affiliation(s)
- Alexandros Giannakis
- Department of Neurology, University of Ioannina, University Campus, Stavrou Niarchou Av., Ioannina, Greece.
| | - Spiridon Konitsiotis
- Department of Neurology, University of Ioannina, University Campus, Stavrou Niarchou Av., Ioannina, Greece
| |
Collapse
|
46
|
Pichet Binette A, Smith R, Salvadó G, Tideman P, Glans I, van Westen D, Groot C, Ossenkoppele R, Stomrud E, Parchi P, Zetterberg H, Blennow K, Mattsson-Carlgren N, Janelidze S, Palmqvist S, Hansson O. Evaluation of the Revised Criteria for Biological and Clinical Staging of Alzheimer Disease. JAMA Neurol 2025:2833818. [PMID: 40388185 DOI: 10.1001/jamaneurol.2025.1100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Importance While clinical disease stages remained largely unchanged in the 2024 update of the Alzheimer disease (AD) criteria, tau-positron emission tomography (PET) was introduced as a core biomarker and its spatial extent was incorporated into the revised biological stages of the disease. It is important to consider both the clinical and the biological stages and understand their discrepancies. Objective To compare individuals who have discrepant biological and clinical stages with those who have congruent stages in terms of copathologies, comorbidities, and demographics. Design, Setting, and Participants Participants were from the Swedish BioFINDER-2 (inclusion from 2017 through 2023) and the Alzheimer's Disease Neuroimaging Initiative (ADNI) (inclusion from 2015 through 2024). BioFINDER-2 included a prospective population-based (cognitively normal [CN] older adults) and memory clinic-based cohort (participants with subjective cognitive impairment [SCD], mild cognitive impairment [MCI], and dementia). ADNI included a volunteer-based sample. All participants who were amyloid-β positive and had undergone tau-PET were included. In BioFINDER-2, 838 participants of a total of 1979 were included, and of 927 with tau-PET in ADNI, 380 were included. Exposures The clinical (CN to dementia) and biological (based on PET; initial [amyloid-β-positive only] to advanced [amyloid-β-positive, elevated, and widespread tau]) stages from the revised AD criteria. Main Outcomes and Measures Cross-sectional measures of neurodegeneration (cortical thickness, TAR DNA-binding protein 43 [TDP-43] imaging signature, neurofilament light [NfL]), α-synuclein cerebrospinal fluid status, plasma glial fibrillary acidic protein, white matter lesions, infarcts, microbleeds, comorbidities, and demographics. Results There were 838 BioFINDER-2 participants (mean age, 73.9 [SD, 7.3] years; 431 women [51%]; 407 men [49%]) and 380 ADNI participants (average age, 72.9 [SD, 7.0] years; 194 women [51%]; 186 mean [49%]) included. In BioFINDER-2, 37.7% of the sample had congruent biological and clinical stages (reference group), 51.3% had more advanced clinical impairment compared with their clinical stage (clinical > biological) and 11.0% had the opposite (biological > clinical). The main differences were between the reference group and the clinical > biological group: the latter participants were more often positive for α-synuclein pathology, had higher NfL levels, greater TDP-43-like atrophy, and higher burden of cerebral small vessel disease lesions (all false discovery rate P < .05). The only difference between the biological > clinical and the reference group was that the former had less neurodegeneration (thicker cortex; all false discovery rate P < .001). The main results were replicated in the independent ADNI cohort, where congruent 56.1% of participants had biological and clinical stages; 36.1% were in the category clinical > biological, and 7.9% in biological > clinical. Conclusions and Relevance Copathologies play an important role in symptom severity in individuals who harbor less tau-tangle pathology than expected for their clinical impairment. These results highlight the importance of measuring non-AD biomarkers in patients with AD with worse cognitive impairment than expected based on their biological stage, which could impact the clinical diagnosis and prognosis.
Collapse
Affiliation(s)
- Alexa Pichet Binette
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Physiology and Pharmacology, Université de Montréal, Montréal, Quebec, Canada
- Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal, Montréal, Quebec, Canada
| | - Ruben Smith
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Gemma Salvadó
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
| | - Pontus Tideman
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Isabelle Glans
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Danielle van Westen
- Diagnostic Radiology, Institute for Clinical Sciences Lund, Lund University, Sweden
- Image and Function, Skåne University Hospital, Lund, Sweden
| | - Colin Groot
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Piero Parchi
- IRCCS, Istituto delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, United Kingdom
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, 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, PR China
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Faculty of Medicine, Lund University, Lund, Sweden
| |
Collapse
|
47
|
Zabel P, Przekoracka-Krawczyk A, Jaworski D, Zabel K, Suwala K, Gebska-Toloczko M, Kucharski R, Kaluzny JJ. Assessment of individual retinal layer thickness and vascular changes in Alzheimer's disease. Sci Rep 2025; 15:17287. [PMID: 40389598 PMCID: PMC12089464 DOI: 10.1038/s41598-025-02377-1] [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: 02/16/2025] [Accepted: 05/13/2025] [Indexed: 05/21/2025] Open
Abstract
Alzheimer's disease (AD) is a leading cause of dementia, underscoring the need for early and accurate diagnostic methods. This cross-sectional study examines the thickness of individual retinal layers and vessel density (VD) in 23 patients with AD and 22 healthy controls (HC). All participants underwent comprehensive ophthalmologic assessments and cognitive evaluations. AD patients exhibited significantly reduced mean macular retinal thickness compared to HC in foveal and parafoveal areas (p < 0.05). The most significant differences were noted in specific sectors of the Inner Nuclear Layer (INL) and Outer Nuclear Layer (ONL) (p < 0.05), while no significant changes were found in the Outer Retinal Layer (p > 0.05). Additionally, AD patients showed decreased VD in the deep vascular plexus (DVP) (p = 0.030) and an increased foveal avascular zone (FAZ) (p = 0.021). The areas under the receiver operating characteristic curves (AUC) analysis revealed the highest diagnostic accuracy for ONL thickness in the S2 sector (0.79) and for mean macular thickness in C0 and T1 sectors (0.76). Positive correlations were observed in some areas between individual retinal layers and VD in their corresponding vascular plexuses in the AD group. The findings suggest that alterations in retinal layer thickness and VD may serve as potential non-invasive diagnostic markers for AD.
Collapse
Affiliation(s)
- Przemyslaw Zabel
- Department of Sensory Organ Studies, Collegium Medicum, Nicolaus Copernicus University, Sandomierska 16, Bydgoszcz, 85-830, Poland.
- Department of Ophthalmology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland.
- Oftalmika Eye Hospital, Bydgoszcz, Poland.
| | - Anna Przekoracka-Krawczyk
- Laboratory of Vision Science and Optometry, Faculty of Physics and Astronomy, Adam Mickiewicz University, Poznan, Poland
| | - Damian Jaworski
- Oftalmika Eye Hospital, Bydgoszcz, Poland
- Division of Ophthalmology and Optometry, Department of Ophthalmology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Katarzyna Zabel
- Department of Sensory Organ Studies, Collegium Medicum, Nicolaus Copernicus University, Sandomierska 16, Bydgoszcz, 85-830, Poland
- Department of Ophthalmology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
- Oftalmika Eye Hospital, Bydgoszcz, Poland
| | - Karolina Suwala
- Department of Sensory Organ Studies, Collegium Medicum, Nicolaus Copernicus University, Sandomierska 16, Bydgoszcz, 85-830, Poland
- Oftalmika Eye Hospital, Bydgoszcz, Poland
| | - Martyna Gebska-Toloczko
- Department of Sensory Organ Studies, Collegium Medicum, Nicolaus Copernicus University, Sandomierska 16, Bydgoszcz, 85-830, Poland
| | | | - Jakub J Kaluzny
- Department of Sensory Organ Studies, Collegium Medicum, Nicolaus Copernicus University, Sandomierska 16, Bydgoszcz, 85-830, Poland
- Oftalmika Eye Hospital, Bydgoszcz, Poland
| |
Collapse
|
48
|
Wartchow KM, Rodrigues L, Dartora WJ, Biasibetti R, Selistre NG, Lazarian A, Barrios-Castellanos C, Bartelo N, Gonçalves CA, McIntire LBJ, on behalf of Alzheimer’s Disease Neuroimaging Initiative (ADNI). Sex-Associated Cerebellar and Hippocampal Volume Reduction in Alzheimer's Disease: Insights from the Clinical ADNI Cohort and STZ Animal Model. Int J Mol Sci 2025; 26:4810. [PMID: 40429951 PMCID: PMC12112559 DOI: 10.3390/ijms26104810] [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: 04/05/2025] [Revised: 05/08/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
While the greatest risk factor for Alzheimer's disease (AD) is aging, women are disproportionately affected by the disease. Interestingly, the hippocampus and cerebellum exhibit gender-specific cytoarchitecture differences, which are associated with AD, despite the absence of a role in animal reproductive behavior or hormonal signaling. This study investigates the potential association of sex differences associated with AD by interrogating cerebellar and hippocampal volume in preclinical (MCI) as well as clinical phases of AD compared to cognitively normal patients (CN) and in an animal model of AD, the streptozotocin (STZ)-induced sporadic AD model. In order to investigate putative changes in cerebellum and hippocampus in a rat model of AD, we used a STZ-induced sporadic AD model at three different time points (2, 4, and 8 weeks) after surgery in male and female rats. Previous studies have reported hippocampal-dependent changes as well as sex-dependent behavioral and signaling effects in the STZ animal model of sporadic AD while our current study showed involvement of cerebellum-mediated changes. To interrogate the role of cerebellar volume in AD progression within the human context, we analyzed data available through the Alzheimer's Disease Neuroimaging Initiative (ADNI). In a cross-sectional analysis, we observed that levels of peripheral Glial Acidic Fibrillary Protein (GFAP) (astrocytic protein) were associated negatively with cerebellar and hippocampal volumes (β = -0.002, p-value = 0.04; β = -6.721, p-value < 0.0001) and were associated with sex specific differences in males. Our analysis identified that the effect on hippocampal volume was earlier in disease stage, reinforcing the relevance of longitudinal alterations of cerebellum and hippocampus volume over time. The STZ animal model of sporadic AD, corroborated the progressive changes in hippocampal volume and more minor and temporally delayed involvement of the cerebellum volume changes which were dependent on sex. This suggests that cerebellar involvement may be secondary to hippocampal neurodegeneration, and both regional differences were dependent on sex. Due to the association with GFAP, our findings may be due to network astrocyte connection spread regardless of primary pathology. Overall, our study uncovers a novel role for cerebellum in AD in a model and in the human context.
Collapse
Affiliation(s)
- Krista Mineia Wartchow
- Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY 10065, USA; (W.J.D.); (A.L.); (C.B.-C.); (N.B.); (L.B.J.M.)
| | - Leticia Rodrigues
- Biochemistry Post-Graduate Program, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, Brazil; (L.R.); (R.B.); (N.G.S.); (C.-A.G.)
| | - William Jones Dartora
- Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY 10065, USA; (W.J.D.); (A.L.); (C.B.-C.); (N.B.); (L.B.J.M.)
| | - Regina Biasibetti
- Biochemistry Post-Graduate Program, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, Brazil; (L.R.); (R.B.); (N.G.S.); (C.-A.G.)
| | - Nicholas Guerini Selistre
- Biochemistry Post-Graduate Program, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, Brazil; (L.R.); (R.B.); (N.G.S.); (C.-A.G.)
| | - Artur Lazarian
- Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY 10065, USA; (W.J.D.); (A.L.); (C.B.-C.); (N.B.); (L.B.J.M.)
| | - Carmen Barrios-Castellanos
- Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY 10065, USA; (W.J.D.); (A.L.); (C.B.-C.); (N.B.); (L.B.J.M.)
| | - Nicholas Bartelo
- Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY 10065, USA; (W.J.D.); (A.L.); (C.B.-C.); (N.B.); (L.B.J.M.)
| | - Carlos-Alberto Gonçalves
- Biochemistry Post-Graduate Program, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre 90035-003, Brazil; (L.R.); (R.B.); (N.G.S.); (C.-A.G.)
| | - Laura Beth J. McIntire
- Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY 10065, USA; (W.J.D.); (A.L.); (C.B.-C.); (N.B.); (L.B.J.M.)
| | | |
Collapse
|
49
|
Perron J, Scramstad C, Ko JH. Brain metabolic imaging-based model identifies cognitive stability in prodromal Alzheimer's disease. Sci Rep 2025; 15:17187. [PMID: 40382421 PMCID: PMC12085605 DOI: 10.1038/s41598-025-02039-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 05/09/2025] [Indexed: 05/20/2025] Open
Abstract
The recent approval of anti-amyloid pharmaceuticals for the treatment of Alzheimer's disease (AD) has created a pressing need for the ability to accurately identify optimal candidates for anti-amyloid therapy, specifically those with evidence for incipient cognitive decline, since patients with mild cognitive impairment (MCI) may remain stable for several years even with positive AD biomarkers. Using fluorodeoxyglucose PET and biomarker data from 594 ADNI patients, a neural network ensemble was trained to forecast cognition from MCI diagnostic baseline. Training data comprised PET studies of patients with biological AD. The ensemble discriminated between progressive and stable prodromal subjects (MCI with positive amyloid and tau) at baseline with 88.6% area-under-curve, 88.6% (39/44) accuracy, 73.7% (14/19) sensitivity and 100% (25/25) specificity in the test set. It also correctly classified all other test subjects (healthy or AD continuum subjects across the cognitive spectrum) with 86.4% accuracy (206/239), 77.4% sensitivity (33/42) and 88.23% (165/197) specificity. By identifying patients with prodromal AD who will not progress to dementia, our model could significantly reduce overall societal burden and cost if implemented as a screening tool. The model's high positive predictive value in the prodromal test set makes it a practical means for selecting candidates for anti-amyloid therapy and trials.
Collapse
Affiliation(s)
- Jarrad Perron
- Graduate Program in Biomedical Engineering, Price Faculty of Engineering, University of Manitoba, 75 Chancellor's Circle, Winnipeg, MB, R3T 5V6, Canada
- PrairieNeuro Research Centre, Kleysen Institute for Advanced Medicine, Health Sciences Centre, 710 William Avenue, Winnipeg, MB, R3E 3J7, Canada
| | - Carly Scramstad
- Section of Neurology, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Ji Hyun Ko
- Graduate Program in Biomedical Engineering, Price Faculty of Engineering, University of Manitoba, 75 Chancellor's Circle, Winnipeg, MB, R3T 5V6, Canada.
- PrairieNeuro Research Centre, Kleysen Institute for Advanced Medicine, Health Sciences Centre, 710 William Avenue, Winnipeg, MB, R3E 3J7, Canada.
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, University of Manitoba, 130-745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada.
| |
Collapse
|
50
|
Yang W, Yu Q, Wang N, Lam KK, Lin ZX, Xian YF. Far-Infrared Radiation Ameliorates the Cognitive Dysfunction in an Alzheimer's Disease Transgenic Mouse via Modulating Jak-2/Stat3 and Nrf-2/HO-1 Pathways. Neuromolecular Med 2025; 27:34. [PMID: 40374872 DOI: 10.1007/s12017-025-08860-2] [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: 04/02/2025] [Accepted: 05/02/2025] [Indexed: 05/18/2025]
Abstract
Alzheimer's disease (AD) is the primary cause of dementia in the elderly. However, effective therapies that modify the disease process in AD remain elusive. Far-infrared radiation (FIR) is commonly utilized as a complementary treatment a range of disease, for example insomnia and rheumatoid arthritis. In this research, we explored how FIR light impacts the cognitive functions of TgCRND8 AD mice and elucidated its underlying molecular mechanism. The cognitive capabilities of TgCRND8 mice assessed by employing the Morris water maze. The concentrations of IL-1β, TNF-α, IL-4, Aβ40, and Aβ42 protein were assessed by enzyme-linked immunosorbent assay. Immunostaining was conducted to assess the Aβ deposits and microglial presence in the brains of TgCRND8 mice. Western blot was applied to detect the protein expressions of tau phosphorylation, amyloid-β (Aβ) production, Jak-2/Stat3, and Nrf-2/HO-1 pathways. The results indicated that FIR light notably ameliorated the cognitive impairments of the AD mice, reduced both Aβ deposition and tau protein hyperphosphorylation at sites of Thr205, Ser369, Ser404, and Thr181, suppressed the release of TNF-α and IL-1β, attenuated the ratios of p-Jak-2/Jak-2 and p-Stat3/Stat3, while increased the protein levels of IL-4, Nrf-2, and HO-1 in the brains of TgCRND8 mice. These findings amply demonstrated that FIR light ameliorated cognitive deficits of TgCRND8 mice via reducing both Aβ burden and tau protein hyperphosphorylation, suppressing the neuroinflammation, and restoring the levels of the oxidative-related proteins through modulating Jak-2/Stat3 and Nrf-2/HO-1 pathways. These experimental findings indicate that FIR light treatment is a promising treatment approach for AD.
Collapse
Affiliation(s)
- Wen Yang
- School of Chinese Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Qiuxia Yu
- School of Chinese Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Nick Wang
- Nick Wang Technology Limited, TML Tower, 3 Hoi Shing Road, Tsuen Wan, Kowloon, Hong Kong SAR, People's Republic of China
| | - Koon Kit Lam
- School of Chinese Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Zhi-Xiu Lin
- School of Chinese Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China.
- Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, People's Republic of China.
| | - Yan-Fang Xian
- School of Chinese Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China.
| |
Collapse
|