1
|
Paitel ER, Pettigrew C, Moghekar A, Miller MI, Faria AV, Albert M, Soldan A. Alzheimer's disease cerebrospinal fluid biomarker levels and APOE genetic status are associated with hippocampal-cerebellar functional connectivity. Neurobiol Aging 2025; 151:107-116. [PMID: 40273528 PMCID: PMC12101073 DOI: 10.1016/j.neurobiolaging.2025.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/20/2025] [Accepted: 04/12/2025] [Indexed: 04/26/2025]
Abstract
Recent research suggests that hippocampal-cerebellar (Hp-CB) functional connectivity may be altered early in the course of Alzheimer's disease (AD), given the early accumulation of AD pathology in the hippocampi and emerging evidence of cerebellar changes in early AD. This study analyzed the role of AD genetic risk (via APOE ε4 carrier status) and cerebrospinal fluid (CSF) biomarkers of AD pathology (ratio of phosphorylated tau (p-tau181) to amyloid beta (Aβ42/Aβ40)) on the relationship between age and functional Hp-CB resting state fMRI connectivity in 161 cognitively unimpaired older adults (M age =67.3; SD =9.0; 37 % APOE ε4 +). In multiple regression analyses with Hp-CB connectivity as the outcome, there were significant interactions between age and APOE ε4 status, and between age and CSF AD biomarkers. Older age was associated with greater Hp-CB connectivity in APOE ε4 non-carriers and participants with less abnormal CSF AD biomarkers. In contrast, Hp-CB connectivity was marginally lower with older age in ε4 carriers and those with more abnormal AD biomarkers. Furthermore, greater Hp-CB connectivity was associated with better episodic memory performance across all groups. These findings suggest that age-related increases in Hp-CB connectivity among APOE ε4 non-carriers and those with low AD biomarker levels reflect age-related changes that are largely unrelated to AD, while age-related decreases in Hp-CB connectivity in APOE ε4 carriers may reflect AD-related alterations. These findings also highlight the importance of cerebellar contributions to cognitive performance among older adults and suggest that Hp-CB connectivity may be altered in preclinical AD.
Collapse
Affiliation(s)
- Elizabeth R Paitel
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Corinne Pettigrew
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael I Miller
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Andreia V Faria
- Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
| | - Marilyn Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anja Soldan
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
2
|
Ahmed FS, Guenther BA, Thompson JL, Lagerstrom L, Robbins MA. Role of light walking pace on cognition: Findings from the Maine-Syracuse Longitudinal Study. APPLIED NEUROPSYCHOLOGY. ADULT 2025; 32:978-992. [PMID: 37402210 PMCID: PMC10764642 DOI: 10.1080/23279095.2023.2228952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 07/06/2023]
Abstract
Moderate- to vigorous intensities of physical activity are recommended for health promotion, including brain health. Regular physical activity is considered a modifiable factor to delay -perhaps prevent- onset of dementias such as Alzheimer's disease. Little is known about the benefits of light physical activity. We analyzed data from a 998 community-dwelling, cognitively unimpaired participants from the Maine-Syracuse Longitudinal Study (MSLS) and investigated the role of light physical activity, defined by walking pace, across two time points. Results revealed light levels of walking pace were associated with higher performance at the first timepoint and less decline by time 2 in the domains of verbal abstract reasoning and visual scanning and tracking, which includes both processing speed and executive function skills. When examining change over time (N = 583), increasing walking pace was associated with less decline at time two for the domains of visual scanning and tracking, working memory, visual spatial ability, and working memory, but not verbal abstract reasoning. These findings highlight the relevance of light physical activity and the need to investigate its contribution to cognitive function. From a public health perspective, this may encourage more adults to adopt a light level of exercise and still reap health benefits.
Collapse
Affiliation(s)
- Fayeza S. Ahmed
- Department of Psychology, University of Maine, 301 Beryl Warner Williams Hall, Orono, ME 04469, USA
| | - Benjamin A. Guenther
- Department of Psychology, University of Maine, 301 Beryl Warner Williams Hall, Orono, ME 04469, USA
| | - Jennifer L. Thompson
- Department of Psychology, University of Maine, 301 Beryl Warner Williams Hall, Orono, ME 04469, USA
| | - Lindsey Lagerstrom
- Department of Psychology, University of Maine, 301 Beryl Warner Williams Hall, Orono, ME 04469, USA
| | - Michael A. Robbins
- Department of Psychology, University of Maine, 301 Beryl Warner Williams Hall, Orono, ME 04469, USA
| |
Collapse
|
3
|
Arruda F, Rosselli M, Mejia Kurasz A, Loewenstein DA, DeKosky ST, Lang MK, Conniff J, Vélez-Uribe I, Ahne E, Shihadeh L, Adjouadi M, Goytizolo A, Barker WW, Curiel RE, Smith GE, Duara R. Stability in cognitive classification as a function of severity of impairment and ethnicity: A longitudinal analysis. APPLIED NEUROPSYCHOLOGY. ADULT 2025; 32:889-902. [PMID: 37395391 DOI: 10.1080/23279095.2023.2222861] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
OBJECTIVE The interaction of ethnicity, progression of cognitive impairment, and neuroimaging biomarkers of Alzheimer's Disease remains unclear. We investigated the stability in cognitive status classification (cognitively normal [CN] and mild cognitive impairment [MCI]) of 209 participants (124 Hispanics/Latinos and 85 European Americans). METHODS Biomarkers (structural MRI and amyloid PET scans) were compared between Hispanic/Latino and European American individuals who presented a change in cognitive diagnosis during the second or third follow-up and those who remained stable over time. RESULTS There were no significant differences in biomarkers between ethnic groups in any of the diagnostic categories. The frequency of CN and MCI participants who were progressors (progressed to a more severe cognitive diagnosis at follow-up) and non-progressors (either stable through follow-ups or unstable [progressed but later reverted to a diagnosis of CN]) did not significantly differ across ethnic groups. Progressors had greater atrophy in the hippocampus (HP) and entorhinal cortex (ERC) at baseline compared to unstable non-progressors (reverters) for both ethnic groups, and more significant ERC atrophy was observed among progressors of the Hispanic/Latino group. For European Americans diagnosed with MCI, there were 60% more progressors than reverters (reverted from MCI to CN), while among Hispanics/Latinos with MCI, there were 7% more reverters than progressors. Binomial logistic regressions predicting progression, including brain biomarkers, MMSE, and ethnicity, demonstrated that only MMSE was a predictor for CN participants at baseline. However, for MCI participants at baseline, HP atrophy, ERC atrophy, and MMSE predicted progression.
Collapse
Affiliation(s)
- Fernanda Arruda
- Department of Psychology, Charles E. Schmidt College of Science, Florida Atlantic University, Davie, FL
| | - Mónica Rosselli
- Department of Psychology, Charles E. Schmidt College of Science, Florida Atlantic University, Davie, FL
- 1Florida Alzheimer's Disease Research Center, Miami Beach and Gainesville, FL, USA
| | - Andrea Mejia Kurasz
- Department of Clinical and Health Psychology, University of Florida College of Public Health and Health Professions, Gainesville, FL, USA
| | - David A Loewenstein
- 1Florida Alzheimer's Disease Research Center, Miami Beach and Gainesville, FL, USA
- Department of Psychiatry and Behavioral Sciences and Center for Cognitive Neuroscience and Aging, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Steven T DeKosky
- 1Florida Alzheimer's Disease Research Center, Miami Beach and Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Merike K Lang
- Department of Psychology, Charles E. Schmidt College of Science, Florida Atlantic University, Davie, FL
| | - Joshua Conniff
- Department of Psychology, Charles E. Schmidt College of Science, Florida Atlantic University, Davie, FL
| | - Idaly Vélez-Uribe
- Department of Psychology, Charles E. Schmidt College of Science, Florida Atlantic University, Davie, FL
- Wien Center for Alzheimer's Disease and Memory Disorders, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Emily Ahne
- Department of Psychology, Charles E. Schmidt College of Science, Florida Atlantic University, Davie, FL
| | - Layaly Shihadeh
- Department of Psychology, Charles E. Schmidt College of Science, Florida Atlantic University, Davie, FL
| | - Malek Adjouadi
- 1Florida Alzheimer's Disease Research Center, Miami Beach and Gainesville, FL, USA
- Center for Advanced Technology and Education, College of Engineering, Florida International University, Miami, FL, USA
| | - Alicia Goytizolo
- Department of Psychology, Charles E. Schmidt College of Science, Florida Atlantic University, Davie, FL
| | - Warren W Barker
- 1Florida Alzheimer's Disease Research Center, Miami Beach and Gainesville, FL, USA
- Wien Center for Alzheimer's Disease and Memory Disorders, Mount Sinai Medical Center, Miami Beach, FL, USA
| | - Rosie E Curiel
- 1Florida Alzheimer's Disease Research Center, Miami Beach and Gainesville, FL, USA
- Department of Psychiatry and Behavioral Sciences and Center for Cognitive Neuroscience and Aging, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Glenn E Smith
- 1Florida Alzheimer's Disease Research Center, Miami Beach and Gainesville, FL, USA
- Department of Clinical and Health Psychology, University of Florida College of Public Health and Health Professions, Gainesville, FL, USA
| | - Ranjan Duara
- 1Florida Alzheimer's Disease Research Center, Miami Beach and Gainesville, FL, USA
- Wien Center for Alzheimer's Disease and Memory Disorders, Mount Sinai Medical Center, Miami Beach, FL, USA
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Dubbelman MA, Liu A, Donohue MC, Langford O, Raman R, Rentz DM, Amariglio R, Sperling RA, Aisen PS, Marshall GA, as the A4 Study team. Changes in Daily Functioning in Association With Tau and Amyloid Among Unimpaired Older Adults With and Without Elevated Amyloid. Neurology 2025; 104:e213775. [PMID: 40440591 PMCID: PMC12123750 DOI: 10.1212/wnl.0000000000213775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 04/02/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Everyday functioning declines gradually over time in Alzheimer disease (AD), with the earliest changes potentially occurring at the preclinical stage. We investigated how changes in everyday functioning relate to (changes in) amyloid and tau in a large sample of cognitively unimpaired older adults, most of whom had elevated amyloid levels at the start of the study. METHODS This prospective study included participants from a 240-week randomized controlled trial of an anti-amyloid drug, solanezumab, and individuals who screen-failed because of a negative amyloid PET scan. A subset (n = 434) underwent repeated tau PET scans. Participants and their study partners completed the Alzheimer's Disease Cooperative Study Activities of Daily Living Prevention Instrument multiple times during the double-blind phase of the trial. Using generalized least-squares models, fit by restricted maximum likelihood, we analyzed how changes in everyday functioning related to amyloid and tau PET. We also correlated changes in amyloid and tau PET with changes in everyday functioning. RESULTS A total of 1,707 participants (71.5 ± 4.7 years, 60% female) showed a marginal decline in everyday functioning. Among individuals with elevated amyloid, those with the highest levels of neocortical and medial temporal tau showed the largest decline in everyday functioning, as reported by the participants and their study partners. Increases in neocortical and medial temporal tau correlated moderately (correlation coefficient ranging from -0.2 to -0.5) with a decline in ADCS ADL-PI scores. Functional changes were not evident with amyloid alone. At the item level, participants and their study partners were most likely to report increased difficulty at the last visit with completing complex activities and selecting and paying for items when shopping. DISCUSSION Higher tau levels are associated with the fastest decline in everyday functioning in the presence of elevated amyloid, and those accumulating more tau show a faster decline in everyday functioning. These findings demonstrate the utility of including sensitive measures of everyday functioning in clinical practice and clinical trials at the stage of preclinical AD. TRIAL REGISTRATION INFORMATION The A4 study, ClinicalTrials.gov ID: NCT02008357; and Longitudinal Evaluation of Amyloid Risk and Neurodegeneration, ClinicalTrials.gov ID: NCT02488720.
Collapse
Affiliation(s)
- Mark A Dubbelman
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School; and
| | - Andy Liu
- Alzheimer Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego, CA
| | - Michael C Donohue
- Alzheimer Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego, CA
| | - Oliver Langford
- Alzheimer Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego, CA
| | - Rema Raman
- Alzheimer Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego, CA
| | - Dorene M Rentz
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School; and
| | - Rebecca Amariglio
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School; and
| | - Reisa Anne Sperling
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School; and
| | - Paul S Aisen
- Alzheimer Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego, CA
| | - Gad A Marshall
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School; and
| | | |
Collapse
|
7
|
Chen X, Thompson J, Yao Z, Cappelleri JC, Amponsah J, Mukherjee R, Hu J. Explainable AI predicting Alzheimer's disease with latent multimodal deep neural networks. J Biopharm Stat 2025:1-15. [PMID: 40528446 DOI: 10.1080/10543406.2025.2511194] [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: 09/18/2024] [Accepted: 05/17/2025] [Indexed: 06/20/2025]
Abstract
PURPOSE Alzheimer's disease (AD) is a neurodegenerative disorder characterized by progressive cognitive decline. We proposed a novel latent multimodal deep learning framework to predict AD cognitive status using clinical, neuroimaging, and genetic data. METHODS Three hundred and twenty-two patients aged between 55 and 92 from the ADNI database were included in the study. Confirmatory Factor Analysis (CFA) was applied to derive the latent scores of AD cognitive impairments as the outcome. A multimodal deep neural network with three modalities, including clinical data, imaging data, and genetic data, was constructed. Attention layers and cross attention layers were added to improve prediction; modality importance scores were calculated for interpretation. Mean Absolute Error (MAE) and Mean Squared Error (MSE) were used to evaluate the model performance. RESULTS The CFA demonstrated good fit to the data. The multimodal neural network of clinical and imaging modalities with attention layers was the best predictive model, with an MAE of 0.330 and an MSE of 0.206. Clinical data contributed the most (35%) to the prediction of AD cognitive status. CONCLUSION Our results demonstrated the attention multimodal model's superior performance in predicting the cognitive impairment of AD, introducing attention layers into the model enhanced the prediction performance.
Collapse
Affiliation(s)
- Xi Chen
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jeffrey Thompson
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS, USA
| | - Zijun Yao
- Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, KS, USA
| | | | - Jonah Amponsah
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS, USA
| | - Rishav Mukherjee
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jinxiang Hu
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
8
|
Qi M, Billheimer J, Chang CCH, Janssen I, Brooks MM, Orchard T, Karlamangla AS, Barinas-Mitchell E, Derby CA, McConnell D, Crawford S, El Khoudary SR. High-density Lipoprotein Over Midlife and Future Cognition in Women: The SWAN HDL Ancillary Study. J Clin Endocrinol Metab 2025; 110:1980-1988. [PMID: 39367567 PMCID: PMC12187125 DOI: 10.1210/clinem/dgae697] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 09/17/2024] [Accepted: 10/02/2024] [Indexed: 10/06/2024]
Abstract
CONTEXT Limited data provides evidence-based insights on the association between the comprehensive metrics of high-density lipoproteins (HDL) and cognitive performance, especially in midlife women for whom the benefit might be the greatest. OBJECTIVE To assess the associations of serum HDL metrics including HDL lipid content [HDL cholesterol, phospholipid (HDL-PL), triglyceride], proteins/subclasses [apolipoprotein A-1 (apoA-1); small, medium, large, total HDL particle (HDL-P); and HDL size], and cholesterol efflux capacity with cognitive performance in midlife women. METHODS This prospective cohort study was conducted among 503 midlife women (1234 observations) from the Study of Women's Health Across the Nation HDL ancillary study. Joint models were applied to examine associations of HDL metrics assessed at midlife (50.2 ± 2.9 years, baseline of the current study) and their changes over midlife (6.1 ± 3.9 years of duration) with subsequent cognitive performance [working memory (Digit Span Backward Test), processing speed (Symbol Digit Modalities Test), and episodic memory immediate and delayed recall (East Boston Memory Test)] assessed repeatedly (maximum 5 times) 1.5 ± 1 years later over 7.72 ± 4.10 years of follow-up. RESULTS Higher total HDL-P and smaller HDL size at midlife were associated with a better subsequent immediate recall, delayed recall, and/or processing speed. Greater increase in HDL-PL, apoA-1, medium HDL-P, and total HDL-P and less increase in HDL size over midlife were associated with a better subsequent immediate and/or delayed recall. CONCLUSION Enhancing specific serum HDL metrics during midlife could be promising in cognitive restoration, particularly memory, the initial and predominant symptom of Alzheimer's disease.
Collapse
Affiliation(s)
- Meiyuzhen Qi
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA 15261, USA
| | - Jeffrey Billheimer
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Chung-Chou H Chang
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA 15261, USA
| | - Imke Janssen
- Department of Preventive Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Maria M Brooks
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA 15261, USA
| | - Trevor Orchard
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA 15261, USA
| | - Arun S Karlamangla
- Division of Geriatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Emma Barinas-Mitchell
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA 15261, USA
| | - Carol A Derby
- Department of Neurology and Department of Epidemiology & Public Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Dan McConnell
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Sybil Crawford
- Tan Chingfen Graduate School of Nursing, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Samar R El Khoudary
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA 15261, USA
| |
Collapse
|
9
|
Wittens MMJ, Sima DM, Brys A, Struyfs H, Niemantsverdriet E, De Roeck E, Bastin C, Benoit F, Bergmans B, Bier JC, de Deyn PP, Deryck O, Hanseeuw B, Ivanoiu A, Picard G, Salmon E, Segers K, Sieben A, Thiery E, Tournoy J, van Binst AM, Versijpt J, Smeets D, Bjerke M, Bellio M, Oxtoby NP, Alexander DC, Ribbens A, Engelborghs S. Independent validation and outlier analysis of EuroPOND alzheimer's disease staging model using ADNI and real-world clinical data. Alzheimers Res Ther 2025; 17:134. [PMID: 40524264 DOI: 10.1186/s13195-025-01788-6] [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: 11/18/2024] [Accepted: 06/08/2025] [Indexed: 06/19/2025]
Abstract
BACKGROUND Event-based modeling (EBM) traces sequential progression of events in complex processes like neurodegenerative diseases, adept at handling uncertainties. This study validated an EBM for Alzheimer's disease (AD) staging designed by EuroPOND, an EU-funded Horizon 2020 project, using research and real-world datasets, a crucial step towards application in multi-center trials. METHODS The training dataset comprised 1737 subjects from ADNI-1/GO/2, using the EuroPOND EBM toolbox. Testing datasets included a research cohort from University of Antwerp (controls, CN (n = 46), subjective cognitive decline, SCD (n = 10), mild cognitive impairment, MCI (n = 47), AD dementia, ADD (n = 16)) and a real-world cohort from 9 Belgian Dementia Council memory clinics (CN (n = 91), SCD (n = 66), (non-amnestic) naMCI (n = 54), aMCI (n = 255), and ADD (n = 220). Biomarkers included: 2 clinical scores (Mini Mental State Examination (MMSE), Rey Auditory Verbal Learning Test (RAVLT)); 3 CSF-biomarkers (Aβ1-42, P-tau181, total-Tau); and 4 magnetic resonance imaging (MRI) biomarkers (volumes of the hippocampi, temporal, parietal, and frontal cortices) computed with icobrain dm. The naMCI and aMCI groups were compared by EBM stage proportions, and the model's effectiveness at patient level was evaluated. RESULTS The research cohort's maximum likelihood event sequence comprised CSF Aβ1-42, P-tau181, T-tau, RAVLT, MMSE, and cortical volumes. The clinical cohort's order was frontal cortex volume, MMSE, and remaining cortical regions. aMCI subjects showed higher staging than naMCI, with 54% in the two most advanced stages compared to 38% in naMCI. In the research cohort, 10 outliers were identified with potential mismatches between assigned stages and clinical or biomarker profiles, with CN (n = 4) and SCD (n = 2) subjects assigned in stage 4, one control in stage 9 with abnormal imaging, and three aMCI cases in stage 0 despite clinical or volumetric signs of impairment. CONCLUSIONS This study highlights the generalizability of EuroPOND's AD EBM model across research and real-world clinical datasets, supporting its use in multi-center trials. aMCI subjects generally reside in more advanced stages than naMCI, who may not necessarily have AD, demonstrating utility for precision recruitment/screening.
Collapse
Affiliation(s)
- Mandy M J Wittens
- Dep. of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Dep. of Neurology, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
- Neuroprotection & Neuromodulation (NEUR) Research Group, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Brussel, 1090, Belgium
| | | | | | - Hanne Struyfs
- Dep. of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | | | - Ellen De Roeck
- Dep. of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Neurology and Memory Clinic, ZAS-Hoge Beuken, Antwerp, Belgium
| | - Christine Bastin
- CRC Human Imaging , GIGA Research, University of Liège, Liège, Belgium
- Fonds de la Recherche Scientifique-FNRS, Liège, Belgium
| | - Florence Benoit
- Geriatrics Department, Brugmann University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Bruno Bergmans
- Neurology Department, AZ St-Jan Brugge, Ghent University and Ghent University Hospital, Bruges, Gent, Belgium
| | - Jean-Christophe Bier
- Neurology Department, H. U. B. - Erasme Hospital, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Peter Paul de Deyn
- Laboratory of Neurochemistry and Behaviour, Experimental Neurobiology unit, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | | | - Bernard Hanseeuw
- WELBIO department, WEL Research Institute, Wavre, 1300, Belgium
- Institute of Neuroscience, Université Catholique de Louvain, Brussels, 1200, Belgium
- Department of Neurology, Clinique Universitaires Saint-Luc, Brussels, 1200, Belgium
| | - Adrian Ivanoiu
- Institute of Neuroscience, Université Catholique de Louvain, Brussels, 1200, Belgium
- Department of Neurology, Clinique Universitaires Saint-Luc, Brussels, 1200, Belgium
| | - Gaëtane Picard
- Department of Neurology, Clinique Saint-Pierre, Ottignies, Belgium
| | - Eric Salmon
- GIGA Cyclotron Research Centre, University of Liege, Liège, Belgium
| | - Kurt Segers
- Neurology & Geriatrics Dpt, Brugmann University Hospital, Van Gehuchtenplein 4, Brussels, 1020, Belgium
| | - Anne Sieben
- Neuropathology lab, IBB-NeuroBiobank BB190113, Born Bunge Institute, Antwerp, Belgium
- Department of Pathology, Antwerp University Hospital - UZA, Antwerp, Belgium
- Laboratory of Neurology, Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Evert Thiery
- Department of Neurology, University Hospital Ghent, Ghent University, Ghent, Belgium
| | - Jos Tournoy
- Gerontology & Geriatrics, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
- Department of Geriatric Medicine, UZ Leuven, Leuven, Belgium
| | - Anne-Marie van Binst
- Radiology department, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Jan Versijpt
- Dep. of Neurology, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
- Neuroprotection & Neuromodulation (NEUR) Research Group, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Brussel, 1090, Belgium
| | | | - Maria Bjerke
- Dep. of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Neuroprotection & Neuromodulation (NEUR) Research Group, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Brussel, 1090, Belgium
- Laboratory of Neurochemistry, Dept of Clinical Chemistry, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Maura Bellio
- Department of Computer Science, UCL Hawkes Institute (Centre for Medical Image Computing), University College London, London, UK
| | - Neil P Oxtoby
- Department of Computer Science, UCL Hawkes Institute (Centre for Medical Image Computing), University College London, London, UK
| | - Daniel C Alexander
- Department of Computer Science, UCL Hawkes Institute (Centre for Medical Image Computing), University College London, London, UK
| | | | - Sebastiaan Engelborghs
- Dep. of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
- Dep. of Neurology, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium.
- Neuroprotection & Neuromodulation (NEUR) Research Group, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, Brussel, 1090, Belgium.
| |
Collapse
|
10
|
Kim JH, Park S, Jung H, Lee EH, Lee ES, Lee JJ, Sohn JH. Comparison of Cognitive Deterioration Between Propofol and Remimazolam Anesthesia in ApoE4 Knock-In Mouse Model. Int J Mol Sci 2025; 26:5718. [PMID: 40565183 DOI: 10.3390/ijms26125718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2025] [Revised: 06/12/2025] [Accepted: 06/13/2025] [Indexed: 06/28/2025] Open
Abstract
Perioperative neurocognitive disorder (PND) is a concern following anesthesia, particularly in individuals at risk for Alzheimer's disease (AD). This study compared the cognitive and pathological effects of propofol and remimazolam in a mouse model with AD following surgery. Five-month-old male ApoE4-KI mice underwent abdominal surgery under either propofol (170 mg/kg) or remimazolam (85 mg/kg) anesthesia. Cognitive function was assessed using the Morris water maze and Y-maze, and neuronal apoptosis and amyloid-beta (Aβ) deposition in the CA3 and dentate gyrus (DG) of the hippocampus were evaluated preoperatively and at 2, 4, and 7 days postoperatively. Both groups showed similar postoperative cognitive functions, with increased relative escape latency at day 2 and decreased relative spontaneous alternation at days 4 and 7. However, the neuropathological analysis revealed that propofol-induced significantly more neuronal death in the CA3 (days 4 and 7) and DG (days 2, 4, and 7), and greater Aβ accumulation in the CA3 (days 2 and 4) and DG (days 2 and 7) compared to remimazolam (p < 0.05). Propofol was associated with more pronounced neuropathologic changes in the hippocampus compared to remimazolam. These findings suggest remimazolam may be a safer anesthetic for patients at risk for neurodegenerative disorders, as it is associated with less severe hippocampal pathology, which is characteristic of AD.
Collapse
Affiliation(s)
- Jong-Ho Kim
- Department of Anesthesiology and Pain Medicine, Chuncheon Sacred Heart Hospital, College of Medicine, Hallym University, Chuncheon 24253, Republic of Korea
- Institute of New Frontier Research, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Songyi Park
- Institute of New Frontier Research, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Harry Jung
- Institute of New Frontier Research, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Eun-Hae Lee
- Institute of New Frontier Research, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Eun-Seo Lee
- Institute of New Frontier Research, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Jae-Jun Lee
- Department of Anesthesiology and Pain Medicine, Chuncheon Sacred Heart Hospital, College of Medicine, Hallym University, Chuncheon 24253, Republic of Korea
- Institute of New Frontier Research, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Jong-Hee Sohn
- Institute of New Frontier Research, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
- Department of Neurology, Chuncheon Sacred Heart Hospital, College of Medicine, Hallym University, Chuncheon 24253, Republic of Korea
| |
Collapse
|
11
|
Yong SJ, Teoh SL, Parhar IS, Soga T, Lim WL, Chew J. Antimicrobial peptides and proteins in Alzheimer's and Parkinson's diseases: implications for biomarker exploration. Rev Neurosci 2025:revneuro-2025-0034. [PMID: 40513579 DOI: 10.1515/revneuro-2025-0034] [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: 03/03/2025] [Accepted: 05/26/2025] [Indexed: 06/16/2025]
Abstract
Biomarkers are necessary tools to validate the diagnosis of Alzheimer's and Parkinson's diseases (AD and PD), especially when clinical symptoms are less apparent in the early stages of diseases. Current biomarkers used in clinical practice rely on brain imaging and cerebrospinal fluid (CSF) levels of amyloid-β (Aβ) peptides and α-synuclein (α-syn) for AD and PD, respectively. However, these diagnostic techniques are not only highly invasive and costly, but they also face limitations in diagnostic accuracy, particularly for preclinical or early stages of diseases. Thus, alternative biomarkers have been sought, preferably those in more accessible and convenient biofluids. Given that antimicrobial peptides and proteins (AMPs) may interact with the neuropathogenesis of AD and PD, AMPs have been recognized as promising candidate biomarkers for AD and PD. Therefore, this review examines the literature on how levels of certain AMPs (lactoferrin, hepcidin, defensin, dermcidin, histatin, cathelicidin L-37, prion protein, amylin or islet amyloid polypeptide, substance P or neurokinin-1, and neuropeptide Y) change in the CSF and more accessible biofluids (serum, plasma, tear, or saliva) in AD and PD patients compared to controls. Based on these findings, this review highlights the advantages, challenges, and future directions of AMP-based biomarkers for AD and PD.
Collapse
Affiliation(s)
- Shin Jie Yong
- Department of Biomedical Sciences, Sir Jeffrey Cheah Sunway Medical School, Faculty of Medical and Life Sciences, Sunway University, Bandar Sunway, 47500, Selangor, Malaysia
| | - Seong Lin Teoh
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Ishwar S Parhar
- Center Initiative for Training International Researchers, University of Toyama, Gofuku, Toyama, 930-8555, Japan
| | - Tomoko Soga
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500, Selangor, Malaysia
| | - Wei Ling Lim
- Department of Biomedical Sciences, Sir Jeffrey Cheah Sunway Medical School, Faculty of Medical and Life Sciences, Sunway University, Bandar Sunway, 47500, Selangor, Malaysia
| | - Jactty Chew
- Department of Biomedical Sciences, Sir Jeffrey Cheah Sunway Medical School, Faculty of Medical and Life Sciences, Sunway University, Bandar Sunway, 47500, Selangor, Malaysia
| |
Collapse
|
12
|
Madhusudhana R, Boyle E, Cen Y. An Overview of Glutaminyl Cyclase as a Promising Drug Target for Alzheimer's Disease. Biomedicines 2025; 13:1467. [PMID: 40564185 DOI: 10.3390/biomedicines13061467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2025] [Revised: 06/06/2025] [Accepted: 06/11/2025] [Indexed: 06/28/2025] Open
Abstract
Alzheimer's disease (AD) has become an increasingly pressing concern for the aging population. Current AD treatments mainly focus on cognitive and neuropsychiatric symptoms-with few FDA-approved treatments targeting disease progression itself. The amyloid cascade hypothesis describes the formation and accumulation of β-amyloid (Aβ) oligomers and plaques as a primary event in AD pathogenesis. This hypothesis has served as the foundation of disease-modifying treatment development over the last decade. Recently, glutaminyl cyclase (QC) has been identified as a potential drug target in the amyloid cascade. QC catalyzes the cyclization of Aβ to form pyroglutamated Aβ (pEAβ). pEAβ acts as the seed for the formation of Aβ plaques, thus preventing the formation of pEAβ via QC inhibition, and offers a promising therapeutic strategy against AD. Here, we offer an overview of the pathway QCI research has followed-from the initial testing of imidazole-based inhibitor scaffolds to QCI structural optimization via pharmacophore identification, Varoglutamstat entering clinical trials, and further avenues of bettering specificity and potency for future QCI development.
Collapse
Affiliation(s)
- Rasajna Madhusudhana
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA
| | - Emily Boyle
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA
| | - Yana Cen
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA
- Center for Drug Discovery, Virginia Commonwealth University, Richmond, VA 23219, USA
| |
Collapse
|
13
|
Yan S, Yun X, Liu Q, Hong Z, Chen Y, Zhang S. Advances in gait research related to Alzheimer's disease. Front Neurol 2025; 16:1548283. [PMID: 40529444 PMCID: PMC12172508 DOI: 10.3389/fneur.2025.1548283] [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: 12/19/2024] [Accepted: 03/27/2025] [Indexed: 06/20/2025] Open
Abstract
Introduction Alzheimer's disease (AD) represents a degenerative condition affecting the nervous system, characterized by the absence of a definitive cause and a lack of a precise therapeutic intervention. Extensive research efforts are being conducted worldwide to enhance early detection methods for AD and to develop medications capable of effectively halting the initiation and progression of the disease during its early stages. Some current detection methods for early diagnosis are expensive and require invasive procedures. More and more evidence shows that gait is related to cognition. A deeper investigation into the intricate interplay between gait and cognition is necessary to elucidate their reciprocal influences and the temporal sequence of these interactions. In the future, it is hoped that with the results of clinical manifestations, neuroimaging, and electrophysiology, simple and objective gait analysis results can be used as an alternative biomarker for cognitive decline to diagnose dementia early. Research objective This research offers a comprehensive scoping review of the contemporary landscape of clinical gait evaluation. It delineates the pertinent concepts of gait analysis and machine learning in AD and elucidates the intricate interplay between gait patterns and cognitive status. Methods A comprehensive literature search was conducted within PubMed for all articles published until march 18, 2024, using a set of keywords, including "machine learning and gait "and "gait and Alzheimer." original articles that met the selection criteria were included. Results and significance A strong correlation exists between autonomous gait and cognitive attributes, necessitating further investigation into the selective interplay between gait and mental factors. Conversely, the gait information of Alzheimer's disease (AD) patients can be captured using a 3D gait analysis system. Numerous gait characteristics can be derived from this gait data, and the early identification of AD can be facilitated by applying a graph neural network-based machine learning approach.
Collapse
Affiliation(s)
- Shuding Yan
- Department of Neurology, The Third School of Clinical Medicine (School of Rehabilitation Medicine) of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xiaoping Yun
- Department of Neurology, China Rehabilitation Research Center (CRRC), Beijing, China
| | - Qiang Liu
- Department of Neurological Rehabilitation, The Affiliated Rehabilitation Hospital of Zhejiang Chinese Medical University (Zhejiang Rehabilitation Medical Center), Hangzhou, Zhejiang, China
| | - Zhenmei Hong
- Department of Neurological Rehabilitation, The Affiliated Rehabilitation Hospital of Zhejiang Chinese Medical University (Zhejiang Rehabilitation Medical Center), Hangzhou, Zhejiang, China
| | - Yufan Chen
- Department of Neurology, The Third School of Clinical Medicine (School of Rehabilitation Medicine) of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Shuijing Zhang
- Department of Neurological Rehabilitation, The Affiliated Rehabilitation Hospital of Zhejiang Chinese Medical University (Zhejiang Rehabilitation Medical Center), Hangzhou, Zhejiang, China
| |
Collapse
|
14
|
Voineskos A, Zhukovsky P, Butters M, Lavretsky H, Brown P, Shimony J, Lenze E, Blumberger D, Flint A, Karp J, Roose S, Dickie E, Felsky D, Nichol G, Al-Dabagh Y, Schoer N, Obiri F, Runk A, Conaty K, Mulsant BH. Brain-cognition relationships and treatment outcome in treatment-resistant late-life depression. RESEARCH SQUARE 2025:rs.3.rs-6340032. [PMID: 40502735 PMCID: PMC12155206 DOI: 10.21203/rs.3.rs-6340032/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/28/2025]
Abstract
Older adults with treatment-resistant depression are at significant risk for cognitive impairment. The relationship between treatment response and cognitive function in this population is not well-established. We examined neural correlates of executive and memory function, and their relationship with prospective treatment outcomes. In the context of a longitudinal biomarker study embedded within a multi-center randomized controlled trial for late-life treatment-resistant depression, 397 participants completed baseline neuropsychological testing, and of these 234 adults successfully completed a baseline MRI scan. Multivariate regressions were used to test for brain-cognition associations between memory and executive function and brain functional connectivity, white matter integrity, and gray matter structure. Further, we employed regularized elastic net regressions to identify biomarkers predicting depression remission (MADRS≤10) in the clinical trial. Among participants who completed neuroimaging better cognition was associated with lower connectivity between components of the default mode and the frontoparietal networks and within the frontoparietal network (multivariate r=0.37, p<0.01). Using diffusion imaging data, lower tract integrity in a distributed set of tracts was associated with poorer executive function (multivariate r=0.27, p<0.05). Additionally, gray matter structure was positively associated with cognition (multivariate r=0.38, p<0.05). Education and better structural brain maintenance but not overall health were associated with better cognition. Ongoing treatment resistance was predicted by poorer cognition and gray matter structure. We identified distinct cross-sectional associations between specific neural circuits and variation in cognitive function in people with treatment-resistant late-life depression. We also found worse cognitive function and gray matter structure predicted ongoing treatment resistance to medication offered in the clinical trial.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Eric Lenze
- Washington University School of Medicine
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
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
|
Lee AL, Hwang E, Hwang J. Exploring the diagnostic potential of EEG theta power and interhemispheric correlation of temporal lobe activities in Alzheimer's Disease through random forest analysis. Comput Biol Med 2025; 192:110248. [PMID: 40359673 DOI: 10.1016/j.compbiomed.2025.110248] [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: 11/25/2024] [Revised: 04/19/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Considering the prevalence of Alzheimer's Disease (AD) among the aging population and the limited means of treatment, early detection emerges as a crucial focus area whereas electroencephalography (EEG) provides a promising diagnostic tool. To date, several studies indicated EEG dataset-based models sporting high diagnostic power in distinguishing patients with AD from healthy controls (HC). However, exploration into which features play a crucial role in the diagnosis remains limited. METHODS This study investigates the diagnostic capabilities of EEG for distinguishing patients with AD from HCs through random forest classification on EEG features. Band power and cross-correlation from the resting state EEG dataset of 22 HCs and 160 patients with AD were calculated using Welch's periodogram and Pearson's correlation, respectively. Welch's t-test was applied to identify features demonstrating significant differences between patients with AD and HCs. Band power and cross-correlation were analyzed using a random forest classifier (RFC) and feature-importance analysis. The importance of feature categories, defined as subsets of features grouped by frequency bands (for band power features) or brain regions (for cross-correlation features), was quantified by calculating their average occurrence across all hyperparameter configurations. RESULT Distinct patterns between the eyes-closed and eyes-open conditions in alpha power were not observed for patients with AD (vs. HC), whereas theta power (4-8 Hz) in all regions was higher in patients with AD (vs. HC)(p<0.05). Interhemispheric cross-correlation in the temporal lobes exhibited the most distinguishable distribution for the cross-correlation dataset. An RFC, exploring 512 models with varied hyperparameters followed by feature-importance analysis based on the mean decrease in impurity, highlighted "theta relative power" and "interhemispheric cross-correlation of channel pairs including temporal channels" as the most important features for distinguishing patients with AD from HCs. RFC on theta-band filtered cross-correlation dataset informed by important features demonstrated the robustness of important features across models with different hyperparameter settings. DISCUSSION The models achieved over 97% accuracy and 100% recall in test sets, although the interpretation of this extraordinarily high accuracy warrants caution due to the small dataset size with high data imbalance and the absence of external validation. This methodology demonstrates the efficacy of EEG-based metrics and machine learning in improving our understanding of EEG characteristics in patients with AD, emphasizing the potential of integrating machine learning techniques into clinical practices.
Collapse
Affiliation(s)
- Ahhyun Lucy Lee
- Department of Life Sciences, POSTECH, Pohang, Gyeongsangbukdo, 37673, Republic of Korea; Research Center, Lablup Inc., Seoul, 06161, Republic of Korea; Department of Brain and Cognitive Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Eunjin Hwang
- Research Center, Lablup Inc., Seoul, 06161, Republic of Korea.
| | - Jeongeun Hwang
- Department of Medical IT Engineering, College of Software Convergence, Soonchunhyang University, Asan, Chungcheongnam-do, 31538, Republic of Korea.
| |
Collapse
|
17
|
Gomez K, Yarmey VR, Mane H, San-Miguel A. Microfluidic and Computational Tools for Neurodegeneration Studies. Annu Rev Chem Biomol Eng 2025; 16:195-216. [PMID: 39813728 DOI: 10.1146/annurev-chembioeng-082223-054547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Understanding the molecular, cellular, and physiological components of neurodegenerative diseases (NDs) is paramount for developing accurate diagnostics and efficacious therapies. However, the complexity of ND pathology and the limitations associated with conventional analytical methods undermine research. Fortunately, microfluidic technology can facilitate discoveries through improved biomarker quantification, brain organoid culture, and small animal model manipulation. Because this technology can increase experimental throughput and the number of metrics that can be studied in concert, it demands more sophisticated computational tools to process and analyze results. Advanced analytical algorithms and machine learning platforms can address this challenge in data generated from microfluidic systems, but they can also be used outside of devices to discern patterns in genomic, proteomic, anatomical, and cognitive data sets. We discuss these approaches and their potential to expedite research discoveries and improve clinical outcomes through ND characterization, diagnosis, and treatment platforms.
Collapse
Affiliation(s)
- Kin Gomez
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA;
| | - Victoria R Yarmey
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA;
- Joint Department of Biomedical Engineering, North Carolina State University, University of North Carolina at Chapel Hill, Raleigh, North Carolina, USA
| | - Hrishikesh Mane
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA;
| | - Adriana San-Miguel
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA;
| |
Collapse
|
18
|
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
|
19
|
Vonk JMJ. Time to align sensitive cognitive assessment with protein biomarkers in Alzheimer's disease. J Neuropsychol 2025; 19:172-175. [PMID: 39829197 PMCID: PMC12166651 DOI: 10.1111/jnp.12413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025]
Affiliation(s)
- Jet M. J. Vonk
- Department of Neurology, Memory and Aging CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| |
Collapse
|
20
|
Bigio B, Lima-Filho RAS, Barnhill O, Sudo FK, Drummond C, Assunção N, Vanderborght B, Beasley J, Young S, Korman A, Jones DR, Sultzer DL, Ferreira ST, Mattos P, Head E, Tovar-Moll F, De Felice FG, Lourenco MV, Nasca C. Sex differences in mitochondrial free-carnitine levels in subjects at-risk and with Alzheimer's disease in two independent study cohorts. Mol Psychiatry 2025; 30:2573-2583. [PMID: 39774493 DOI: 10.1038/s41380-024-02862-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 11/14/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025]
Abstract
A major challenge in the development of more effective therapeutic strategies for Alzheimer's disease (AD) is the identification of molecular mechanisms linked to specific pathophysiological features of the disease. Importantly AD has a two-fold higher incidence in women than men and a protracted prodromal phase characterized by amnestic mild-cognitive impairment (aMCI) suggesting that biological processes occurring early can initiate vulnerability to AD. Here, we used a sample of 125 subjects from two independent study cohorts to determine the levels in plasma (the most accessible specimen) of two essential mitochondrial markers acetyl-L-carnitine (LAC) and its derivative free-carnitine motivated by a mechanistic model in rodents in which targeting mitochondrial metabolism of LAC leads to the amelioration of cognitive function and boosts epigenetic mechanisms of gene expression. We report a sex-specific deficiency in free-carnitine levels in women with aMCI and early-AD compared to cognitively healthy controls; no change was observed in men. We also replicated the prior finding of decreased LAC levels in both women and men with AD, supporting the robustness of the study samples assayed in our new study. The magnitude of the sex-specific free-carnitine deficiency reflected the severity of cognitive dysfunction and held in two study cohorts. Furthermore, patients with the lower free-carnitine levels showed higher β-amyloid(Aβ) accumulation and t-Tau levels assayed in cerebrospinal fluid (CSF). Computational analyses showed that the mitochondrial markers assayed in plasma are at least as accurate as CSF measures to classify disease status. Together with the mechanistic platform in rodents, these translational findings lay the groundwork to create preventive individualized treatments targeting sex-specific changes in mitochondrial metabolism that may be subtle to early cognitive dysfunction of AD risk.
Collapse
Affiliation(s)
- Benedetta Bigio
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, USA
| | | | - Olivia Barnhill
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA
| | - Felipe K Sudo
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, RJ, Brazil
| | - Claudia Drummond
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, RJ, Brazil
- Department of Speech and Hearing Pathology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Naima Assunção
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, RJ, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Bart Vanderborght
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, RJ, Brazil
| | - James Beasley
- Biochemical Genetics Laboratory, Duke University Health System, Durham, NC, USA
| | - Sarah Young
- Biochemical Genetics Laboratory, Duke University Health System, Durham, NC, USA
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Aryeh Korman
- Metabolomics Laboratory, NYU Grossman School of Medicine, New York, NY, USA
| | - Drew R Jones
- Metabolomics Laboratory, NYU Grossman School of Medicine, New York, NY, USA
| | - David L Sultzer
- Department of Psychiatry and Human Behavior, School of Medicine, and Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, Irvine, CA, USA
| | - Sergio T Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Rio de Janeiro, RJ, Brazil
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, RJ, Brazil
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Paulo Mattos
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, RJ, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Institute of Psychiatry, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Elizabeth Head
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California Irvine, Irvine, CA, USA
- Department of Pathology and Laboratory Medicine, Department of Neurology, University of California Irvine, Irvine, CA, USA
| | - Fernanda Tovar-Moll
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, RJ, Brazil
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Rio de Janeiro, RJ, Brazil
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, RJ, Brazil
- Centre for Neurosciences Studies, Departments of Biomedical and Molecular Sciences & Department of Psychiatry, Queen's University, Kingston, ON, Canada
| | - Mychael V Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Rio de Janeiro, RJ, Brazil.
| | - Carla Nasca
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA.
- Center for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, USA.
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA.
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
21
|
Vanderlip CR, Stark CEL. Integrating plasma p-tau217 and digital cognitive assessments for early detection in Alzheimer's disease. Alzheimers Dement 2025; 21:e70355. [PMID: 40491259 PMCID: PMC12149436 DOI: 10.1002/alz.70355] [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: 03/04/2025] [Revised: 04/17/2025] [Accepted: 05/15/2025] [Indexed: 06/11/2025]
Abstract
INTRODUCTION Plasma phosphorylated tau (p-tau)217 is an early Alzheimer's disease (AD) biomarker, but the timing of pathological changes and cognitive decline varies substantially. Digital cognitive assessments can detect subtle cognitive changes, suggesting they may complement p-tau217 for early detection. Here, we evaluate whether combining these tools improves the detection of individuals at risk for future decline. METHODS We analyzed 954 amyloid-positive cognitively unimpaired individuals who completed a digital cognitive assessment and a blood test for p-tau217, assessing their ability to predict future decline on the Preclinical Alzheimer Cognitive Composite (PACC) and Mini-Mental State Examination (MMSE). RESULTS Combining performance on a digital cognitive assessment with p-tau217 improved identification of individuals who declined on the PACC and MMSE in the next 5 years. The predictive value was stronger in apolipoprotein E ε4 noncarriers but did not differ by sex. DISCUSSION This approach offers a sensitive method for identifying individuals at high risk for AD-related cognitive decline. HIGHLIGHTS Combining plasma phosphorylated tau 217 with baseline digital cognitive assessment improved the prediction of cognitive decline on gold-standard neuropsychological tests over the next 5 years, achieving greater accuracy than either measure alone. This combination also predicted a decline in a global cognitive screening test. Pairing a blood test with a digital cognitive assessment offers a scalable and feasible approach for Alzheimer's disease screening.
Collapse
Affiliation(s)
- Casey R. Vanderlip
- Department of Neurobiology and BehaviorUniversity of California IrvineIrvineCaliforniaUSA
| | - Craig E. L. Stark
- Department of Neurobiology and BehaviorUniversity of California IrvineIrvineCaliforniaUSA
| |
Collapse
|
22
|
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
|
23
|
Yusup N, Rahmat A, Li H. A preliminary study of the reliability and validity of the Uyghur version of the NUCOG cognitive screening application. BMC Neurol 2025; 25:229. [PMID: 40442633 PMCID: PMC12121169 DOI: 10.1186/s12883-025-04160-1] [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: 02/18/2025] [Accepted: 03/26/2025] [Indexed: 06/02/2025] Open
Abstract
INTRODUCTION Technological advances and artificial intelligence now make it feasible to administer cognitive assessments on touch-screen devices. The aim of this study is to develop a Uyghur version of the NUCOG cognitive screening application and evaluate its reliability, validity, and optimal cutoff scores among Uyghur people with cognitive impairment. METHODS The English version of the NUCOG app was translated and adapted into the Uyghur version (NUCOG-U). A total of 250 Uyghur people aged 55-80, including 90 normal controls, 91 patients with mild cognitive impairment (MCI), and 69 dementia patients, were randomly selected and administered with NUCOG, MoCA-U, Mini-Mental State Examination (MMSE), and other neuropsychological batteries. ROC curves were generated to determine the optimal cutoff values. RESULTS NUCOG-U version showed high internal consistency (Cronbach's α = 0. 826), inter-rater reliability (ICC = 0.999), and test - retest reliability (r = 0.998, p < 0.001). NUCOG scores were significantly correlated with those of MoCA-U (r = 0.896, p < 0.001) and MMSE(r = 0.899, p < 0.001). NUCOG scores were significantly different among the three groups (p < 0.001). The optimal cutoff value for MCI was 80.5, with a sensitivity of 100% and specificity of 73%, and 70 for dementia, with a sensitivity of 94.1% and specificity of 100%. CONCLUSION The NUCOG-U shows high reliability and validity and is suitable for screening cognitive function in the elderly Uyghur population. The optimal cutoff scores to detect mild cognitive impairment and dementia in the Uyghur people are 80.5 and 70, respectively.
Collapse
Affiliation(s)
- Nazuk Yusup
- Department of Neurology, People's Hospital of Xinjiang Uyghur Autonomous Region, Xinjiang Clinical Research Center for Stroke and Neurological Rare Disease, Xinjiang National Center for Cognitive Disorders, Urumqi, 830001, China
| | - Altunsa Rahmat
- Department of Neurology, People's Hospital of Xinjiang Uyghur Autonomous Region, Xinjiang Clinical Research Center for Stroke and Neurological Rare Disease, Xinjiang National Center for Cognitive Disorders, Urumqi, 830001, China
| | - Hongyan Li
- Department of Neurology, People's Hospital of Xinjiang Uyghur Autonomous Region, Xinjiang Clinical Research Center for Stroke and Neurological Rare Disease, Xinjiang National Center for Cognitive Disorders, Urumqi, 830001, China.
| |
Collapse
|
24
|
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 PMCID: PMC12152440 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] [Download PDF] [Figures] [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
|
25
|
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
|
26
|
Losinski GM, Key MN, Vidoni ED, Clutton J, Morris JK, Burns JM, Watts A. APOE4 and chronic health risk factors are associated with sex-specific preclinical Alzheimer's disease neuroimaging biomarkers. Front Glob Womens Health 2025; 6:1531062. [PMID: 40444147 PMCID: PMC12119584 DOI: 10.3389/fgwh.2025.1531062] [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: 11/19/2024] [Accepted: 04/28/2025] [Indexed: 06/02/2025] Open
Abstract
Introduction Two thirds of Alzheimer's disease (AD) patients are female. Genetic and chronic health risk factors for AD affect females more negatively compared to males. Objective This multimodal neuroimaging study aimed to examine sex differences in cognitively unimpaired older adults on: (1) amyloid-β via 18F-AV-45 Florbetapir PET imaging, (2) neurodegeneration via T1 weighted MRI volumetrics, (3) cerebral blood flow via ASL-MRI. We identified AD risk factors including genetic (APOE genotype status) and health markers (fasting glucose, mean arterial pressure, waist-to-hip ratio, and android and gynoid body fat) associated with neuroimaging outcomes for which we observed sex differences. Methods Participants were sedentary, amyloid-β positive older adults (N = 112, ages 65-87 years) without evidence of cognitive impairment (CDR = 0). Results Multivariate analysis of covariance models adjusted for intracranial volume, age, and years of education demonstrated lower volume [F (7, 102) = 2.67, p = 0.014] and higher blood flow F (6, 102) = 4.25, p ≤ 0.001) among females compared to males in regions of interest connected to AD pathology and the estrogen receptor network. We did not observe sex differences in amyloid-β levels. Higher than optimal waist to hip ratio was most strongly associated with lower volume among female participants. Discussion Findings suggest genetic and chronic health risk factors are associated with sex-specific AD neuroimaging biomarkers. Underlying sex-specific biological pathways may explain these findings. Our results highlight the importance of considering sex differences in neuroimaging studies and when developing effective interventions for AD prevention and risk reduction.
Collapse
Affiliation(s)
- Genna M. Losinski
- Department of Psychology, University of Kansas, Lawrence, KS, United States
| | - Mickeal N. Key
- Department of Neurology, University of Kansas Alzheimer’s Disease Center, University of Kansas Medical Center, Fairway, KS, United States
| | - Eric D. Vidoni
- Department of Neurology, University of Kansas Alzheimer’s Disease Center, University of Kansas Medical Center, Fairway, KS, United States
| | - Jonathan Clutton
- Department of Neurology, University of Kansas Alzheimer’s Disease Center, University of Kansas Medical Center, Fairway, KS, United States
| | - Jill K. Morris
- Department of Neurology, University of Kansas Alzheimer’s Disease Center, University of Kansas Medical Center, Fairway, KS, United States
| | - Jeffrey M. Burns
- Department of Neurology, University of Kansas Alzheimer’s Disease Center, University of Kansas Medical Center, Fairway, KS, United States
| | - Amber Watts
- Department of Psychology, University of Kansas, Lawrence, KS, United States
- Department of Neurology, University of Kansas Alzheimer’s Disease Center, University of Kansas Medical Center, Fairway, KS, United States
| |
Collapse
|
27
|
Paris A, Amirthalingam G, Karania T, Foote IF, Dobson R, Noyce AJ, Marshall CR, Waters S. Depression and dementia: interrogating the causality of the relationship. J Neurol Neurosurg Psychiatry 2025; 96:573-581. [PMID: 39798961 DOI: 10.1136/jnnp-2024-334675] [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: 07/16/2024] [Accepted: 11/17/2024] [Indexed: 01/15/2025]
Abstract
BACKGROUND Depression is often cited as a major modifiable risk factor for dementia, though the relative contributions of a true causal relationship, reverse causality and confounding factors remain unclear. This study applied a subset of the Bradford Hill criteria for causation to depression and dementia including strength of effect, specificity, temporality, biological gradient and coherence. METHODS A total of 491 557 participants in UK Biobank aged between 40 and 69 at enrolment and followed up for a mean duration of 12.4 years were studied. Diagnoses of depression and dementia were ascertained from linked health records, self-reports and death certificate registration. Depressive symptoms were measured at enrolment using a combination of questions based on the Patient Health Questionnaire-9 depression screening questionnaire. Regional grey matter volumes were measured using T1-weighted MRI in 41 929 participants. RESULTS Depression was a strong risk factor for incident dementia with an OR of 1.76 (95% CI 1.63 to 1.90), a relationship which was found to be specific to depression rather than commonly proposed confounders. Depressive symptoms increased rapidly in the 10 years prior to dementia diagnosis. The severity of depressive symptoms showed a dose-response relationship with dementia risk. Depression at older ages correlated with reduced grey matter volume in an Alzheimer's pattern whereas younger onset depression was associated with reduced grey matter volume in the frontal lobes and cerebellum. CONCLUSIONS This study provides evidence that the link between depression and dementia is due to reverse causation with a smaller component of causation with clear evidence of both mechanisms driving the association.
Collapse
Affiliation(s)
- Alvar Paris
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Guru Amirthalingam
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Tasvee Karania
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Isabelle F Foote
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, Colorado, USA
| | - Ruth Dobson
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
- Department of Neurology, Barts Health NHS Trust, London, UK
| | - Alastair J Noyce
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
- Department of Neurology, Barts Health NHS Trust, London, UK
| | - Charles R Marshall
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
- Department of Neurology, Barts Health NHS Trust, London, UK
| | - Sheena Waters
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| |
Collapse
|
28
|
Thomas KR, Edmonds EC. Objective Criteria for Subtle Cognitive Decline in Aging and Preclinical Alzheimer Disease: A Systematic Review. Neurology 2025; 104:e213536. [PMID: 40198863 PMCID: PMC11995284 DOI: 10.1212/wnl.0000000000213536] [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: 10/24/2024] [Accepted: 02/04/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Although the Alzheimer's Association (AA) biomarker-only Alzheimer disease (AD) criteria and the International Working Group clinical-pathologic AD criteria differ, both approaches appreciate the need for early detection efforts. Within the AA approach, clinical stage 2 recognizes that someone can be cognitively unimpaired but still experience "subtle cognitive decline" (SCD) measured by subjective report or objective decline using neuropsychological measures. While significant attention has been given to subjective cognitive decline methods, there are no systematic examinations of the operational definition of SCD using objective neuropsychological measures. Therefore, the primary aim of this review was to identify and describe the approaches used to classify objective SCD. METHODS A systematic literature search was performed using PubMed/MEDLINE, Web of Science, and PsycInfo databases for articles with dates ranging from the start of the database through November 1, 2023. Included studies were peer-reviewed, described a discrete objective SCD category, included participants aged 50+ without mild cognitive impairment (MCI) or dementia, and focused on aging or AD. A modified Newcastle-Ottawa Scale was used to assess the quality of included studies. Data were extracted by the 2 authors who then categorized and described the classification approaches. RESULTS Of the 1,361 publications initially identified, 70 case-control studies met criteria for inclusion. SCD definitions generally fell into 6 categories based on using similar methodology: (1) SCD based on a specified cutoff on a single cognitive test (n = 6); (2) SCD based on a cutoff (e.g., 10th percentile) on a cognitive composite score (n = 9); (3) objectively defined SCD (Obj-SCD) using cutoffs (e.g., -1 SD) on multiple individual neuropsychological measures (n = 24); (4) "Pre-MCI" criteria defined using a Clinical Dementia Rating of 0.5 but normal performance on neuropsychological testing (n = 12); (5) cutoff based on longitudinal rate of cognitive decline (e.g., over 1 year) (n = 13); and (6) data-driven/clustering approach to classification (n = 8). Two studies used multiple classification approaches. DISCUSSION Six promising classification approaches were identified in the existing literature, with the Obj-SCD and Pre-MCI approaches being the most commonly applied. Additional work is needed to compare SCD approaches head-to-head to identify the most prognostically useful, particularly within racially/ethnically diverse older adults.
Collapse
Affiliation(s)
- Kelsey R Thomas
- Department of Psychiatry, University of California, San Diego, La Jolla
- Research Service, VA San Diego Healthcare System, CA
| | - Emily C Edmonds
- Banner Alzheimer's Institute, Tucson, AZ; and
- Departments of Neurology and Psychology, University of Arizona, Tucson
| |
Collapse
|
29
|
Lojo-Ramírez JA, Fernández-Rodríguez P, Guerra-Gómez M, Marín-Cabañas AM, Franco-Macías E, Jiménez-Hoyuela-García JM, García-Solís D. Evaluation of early-phase 18F-Florbetaben PET as a surrogate biomarker of neurodegeneration: In-depth comparison with 18F-FDG PET at group and single patient level. J Alzheimers Dis 2025:13872877251340380. [PMID: 40340729 DOI: 10.1177/13872877251340380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
BackgroundImaging biomarkers are essential in Alzheimer's disease (AD) diagnosis, particularly since the introduction of the ATN criteria by the NIA-AA. These criteria include amyloid-β plaques (amyloid PET), fibrillar tau (tau PET), and neurodegeneration (FDG PET or MRI). Early-phase amyloid PET imaging has shown a strong correlation with FDG PET at the group level.ObjectiveThis study evaluates the comparability of early-phase FBB PET (eFBB) perfusion imaging and FDG PET metabolic imaging at both group and individual levels.MethodsA retrospective study included 103 patients with mild cognitive impairment (MCI) or mild dementia suspected of AD who underwent FDG PET and dual-phase 18F-Florbetaben PET (including a 5-min eFBB scan) between 2019 and 2023, along with 33 healthy controls. Imaging analyses included qualitative, semi-quantitative, and voxel-wise techniques to compare eFBB and FDG PET scans.ResultseFBB and FDG PET SUVR values showed a strong correlation across all brain regions (rho = 0.879). Visual assessments of eFBB and FDG PET by two raters achieved intra-observer agreement rates of 87.5% and 86.4%, respectively. Voxel-wise analysis revealed moderate to good overlap, as indicated by Dice-Sørensen coefficients, in the MCI and mild dementia groups. Discriminative performance between eFBB and FDG PET was comparable, with no significant differences as eFBB reliably reflected brain metabolic patterns observed on FDG PET, supporting its diagnostic utility.ConclusionseFBB PET could serve as a surrogate biomarker for FDG PET in the diagnostic evaluation of neurodegenerative dementias. Dual-phase amyloid PET enables simultaneous assessment of neurodegeneration and amyloid burden, offering a streamlined and resource-efficient approach for clinical practice.
Collapse
Affiliation(s)
- Jose Antonio Lojo-Ramírez
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, España
- Servicio de Medicina Nuclear, Hospital Universitario Virgen del Rocío, Sevilla, España
| | - Paula Fernández-Rodríguez
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, España
- Servicio de Medicina Nuclear, Hospital Universitario Virgen del Rocío, Sevilla, España
| | - Miriam Guerra-Gómez
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, España
- Servicio de Medicina Nuclear, Hospital Universitario Virgen del Rocío, Sevilla, España
| | - Alba Marta Marín-Cabañas
- Unidad de Memoria, Servicio de Neurología, Hospital Universitario Virgen del Rocío, Sevilla, España
| | - Emilio Franco-Macías
- Unidad de Memoria, Servicio de Neurología, Hospital Universitario Virgen del Rocío, Sevilla, España
| | - Jose Manuel Jiménez-Hoyuela-García
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, España
- Servicio de Medicina Nuclear, Hospital Universitario Virgen del Rocío, Sevilla, España
| | - David García-Solís
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, España
- Servicio de Medicina Nuclear, Hospital Universitario Virgen del Rocío, Sevilla, España
| |
Collapse
|
30
|
Galal A, Moustafa A, Salama M. Transforming neurodegenerative disorder care with machine learning: Strategies and applications. Neuroscience 2025; 573:272-285. [PMID: 40120712 DOI: 10.1016/j.neuroscience.2025.03.036] [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: 01/21/2025] [Revised: 03/05/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Neurodegenerative diseases (NDs), characterized by progressive neuronal degeneration and manifesting in diverse forms such as memory loss and movement disorders, pose significant challenges due to their complex molecular mechanisms and heterogeneous patient presentations. Diagnosis often relies heavily on clinical assessments and neuroimaging, with definitive confirmation frequently requiring post-mortem autopsy. However, the emergence of Artificial Intelligence (AI) and Machine Learning (ML) offers a transformative potential. These technologies can enable the development of non-invasive tools for early diagnosis, biomarker identification, personalized treatment strategies, patient subtyping and stratification, and disease risk prediction. This review aims to provide a starting point for researchers, both with and without clinical backgrounds, who are interested in applying ML to NDs. We will discuss available data resources for key diseases like Alzheimer's and Parkinson's, explore how ML can revolutionize neurodegenerative care, and emphasize the importance of integrating multiple high-dimensional data sources to gain deeper insights and inform effective therapeutic strategies.
Collapse
Affiliation(s)
- Aya Galal
- Systems Genomics Laboratory, American University in Cairo, New Cairo, Egypt; Institute of Global Health and Human Ecology, American University in Cairo, New Cairo, Egypt
| | - Ahmed Moustafa
- Systems Genomics Laboratory, American University in Cairo, New Cairo, Egypt; Institute of Global Health and Human Ecology, American University in Cairo, New Cairo, Egypt; Biology Department, American University in Cairo, New Cairo, Egypt
| | - Mohamed Salama
- Institute of Global Health and Human Ecology, American University in Cairo, New Cairo, Egypt; Global Brain Health Institute (GBHI), Trinity College Dublin, Dublin 2, Ireland; Faculty of Medicine, Mansoura University, El Mansura, Egypt.
| |
Collapse
|
31
|
Azmoun S, Lewis F, Shoieb D, Jin Y, Colicino E, Winters I, Gu H, Krishnamurthy H, Richardson J, Placidi D, Lambertini L, Lucchini RG. Impact of Manganese on Neuronal Function: An Exploratory MultiOmic Study on Ferroalloy Workers in Brescia, Italy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.05.02.25326824. [PMID: 40385409 PMCID: PMC12083615 DOI: 10.1101/2025.05.02.25326824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Interest is growing in the potential role of manganese (Mn) in Alzheimer's Disease (ADRD). This nested pilot study of a ferroalloy workers cohort was aimed to investigate the effects of long-term occupational Mn exposure on cognitive function through β-amyloid (Aβ) modification and brain deposition, as well as metabolomic, lipidomic and proteomic profiling. We examined 6 male exposed workers (median age 63, exposure duration 31 yrs), and 5 historical controls (median age 60) who had undergone brain PET scan imaging showing higher Aβ deposition among the exposed compared to the controls (p < 0.05). The average annual cumulative respirable Mn of the ferroalloy workers was 329.23 ± 516.39 μg/m3 (geometric mean 118.59). Average Mn level in plasma of the exposed subjects (0.704 ± 0.2 ng/mL) was significantly higher than the controls (0.397 ± 0.18). Pathway analyses using LC-MS/MS results revealed impacted metabolomic pathways such as olfactory signaling, mitochondrial fatty acid beta-oxidation, biogenic amine synthesis, SLC-mediated transmembrane transport, and glycerophospholipid and choline metabolism in the Mn exposed group. Single molecule arrays (Simoa) analysis revealed notable modifications of AD-related plasma biomarkers; protein microarray (chip) showed significant changes (p < 0.05) in the levels of some plasma antibodies targeting autoimmune and neuronal associated proteins such as Aβ (25-35), GFAP, Serotonin, Human NOVA1, and Human Siglec-1/CD169 among the Mn exposed individuals. This data provides evidence on Mn-induced alterations of pathways and biomarkers associated with cognitive neurodegenerative diseases.
Collapse
Affiliation(s)
- Somaiyeh Azmoun
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social work, Florida International University, Miami, FL, USA
| | - Freeman Lewis
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social work, Florida International University, Miami, FL, USA
| | - Daniel Shoieb
- Department of Medical and Surgical Specialties, University of Brescia, Brescia, Italy
| | - Yan Jin
- St. Jude Children Research Hospital, Memphis, TN, USA
| | - Elena Colicino
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Isha Winters
- Isakson Center for Neurological Disease Research and Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, GA, USA
| | - Haiwei Gu
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
| | | | - Jason Richardson
- Isakson Center for Neurological Disease Research and Department of Physiology and Pharmacology, College of Veterinary Medicine, University of Georgia, GA, USA
| | - Donatella Placidi
- Department of Medical and Surgical Specialties, University of Brescia, Brescia, Italy
| | - Luca Lambertini
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Roberto G Lucchini
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social work, Florida International University, Miami, FL, USA
- Occupational Medicine, University of Modena and Reggio Emilia, Italy
| |
Collapse
|
32
|
Ito K, Washimi Y, Kato T, Suzuki K, Ouchi Y, Watanabe C, Sunada Y, Kutoku Y, Ishii K, Ishii K, Kitayama M, Matsubara E, Kimura N, Takano H, Adachi H, Hara K, Kawarabayashi T, Shoji M, Sugimoto N. 18F-FDG PET for the differential diagnosis of Alzheimer's disease and frontotemporal lobar degeneration: A multicenter prospective study in Japan. J Alzheimers Dis 2025:13872877251338691. [PMID: 40325872 DOI: 10.1177/13872877251338691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Background18F-fluoro-2-deoxy-2-D-glucose positron emission tomography (18F-FDG PET) is a biomarker of neuronal injury, according to the revised National Institute on Aging-Alzheimer's Association criteria.ObjectiveThis multicenter prospective cohort study aimed to evaluate the value of 18F-FDG PET for differential diagnosis of Alzheimer's disease (AD) and frontotemporal lobar degeneration (FTLD) in comparison with phosphorylated tau protein (p-tau181) in cerebrospinal fluid (CSF).MethodsIn total, 138 patients (AD, 119; FTLD, 19) from 11 participating institutions underwent clinical and neuropsychological examinations, magnetic resonance imaging (MRI), CSF examination, and 18F-FDG PET at baseline. The cases were visually classified into predefined dementia patterns using 18F-FDG PET by three experts. A region-of-interest (ROI)-based automated analysis of 18F-FDG PET was also performed. The participants were followed up for 12 months, and the clinical diagnosis of dementia was re-evaluated.ResultsThe sensitivity, specificity, and accuracy of the visual reading of18 F-FDG PET for differentiating AD from FTLD were 94%, 78%, and 92%, respectively. In contrast, those of p-tau181 in CSF were 62%, 79%, and 65%, respectively. The sensitivity, the primary endpoint, was 32% higher for 18F-FDG PET than for p-tau181 in CSF. Additionally, the accuracy, the secondary endpoint, was 27% higher for 18F-FDG PET than for p-tau181 in CSF. In addition to the visual reading of 18F-FDG PET, the ROI-based automated analysis showed sensitivity, specificity, and accuracy of 81%, 79%, and 81%, respectively.ConclusionsThis study showed that the diagnostic performance of 18F-FDG PET in differential diagnosis of AD and FTLD was higher than that of p-tau181 in CSF.Trial registrationUMIN-CTR (UMIN 000016427, https://www.umin.ac.jp/ctr/) and Japan Registry of Clinical Trials (jRCTs041180098, https://jrct.mhlw.go.jp/).
Collapse
Affiliation(s)
- Kengo Ito
- National Hospital for Geriatric Medicine, National Centre for Geriatrics and Gerontology, Obu, Japan
| | - Yukihiko Washimi
- National Hospital for Geriatric Medicine, National Centre for Geriatrics and Gerontology, Obu, Japan
| | - Takashi Kato
- National Hospital for Geriatric Medicine, National Centre for Geriatrics and Gerontology, Obu, Japan
| | - Keisuke Suzuki
- National Hospital for Geriatric Medicine, National Centre for Geriatrics and Gerontology, Obu, Japan
| | - Yasuomi Ouchi
- Department of Biofunctional Imaging, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Chigusa Watanabe
- Department of Neurology, National Hospital Organization, Hiroshima-Nishi Medical Center, Otake, Japan
| | - Yoshihide Sunada
- Department of Neurology, Kawasaki Medical School, Kurashiki, Japan
| | - Yumiko Kutoku
- Department of Neurology, Kawasaki Medical School, Kurashiki, Japan
| | - Kazunari Ishii
- Department of Radiology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Kenji Ishii
- Team for Neuroimaging Research, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Michio Kitayama
- Department of Neurology, Okayama Kyokuto Hospital, Okayama, Japan
| | - Etsuro Matsubara
- Department of Neurology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Noriyuki Kimura
- Department of Neurology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Harumasa Takano
- Department of Clinical Neuroimaging, Integrative Brain Imaging Center, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Hiroaki Adachi
- Department of Neurology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kazuhiro Hara
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takeshi Kawarabayashi
- Department of Neurology, Dementia Research Center, Geriatrics Research Institute and Hospital, Maebashi, Japan
| | - Mikio Shoji
- Department of Neurology, Dementia Research Center, Geriatrics Research Institute and Hospital, Maebashi, Japan
| | | |
Collapse
|
33
|
Lee S, Baratono S, Burt G, Palm S, Drew W, Zide B, Chiulli N, Lariviere S, Fox M, Sperling R, Donovan N, Siddiqi S. Localization of Network-Level Atrophy in Preclinical Alzheimer's Disease. RESEARCH SQUARE 2025:rs.3.rs-5977523. [PMID: 40386416 PMCID: PMC12083676 DOI: 10.21203/rs.3.rs-5977523/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
Brain atrophy may precede symptoms in Alzheimer's disease (AD), but it remains unclear whether atrophy in this preclinical stage falls within a distinct brain network or is associated with transitional cognitive changes. We investigated cortical thickness in cognitively unimpaired older adults with varying amyloid-β accumulation and estimated the connectivity of each individual's atrophy pattern using a large normative connectome (n = 1000). A distinct network was connected to atrophy patterns in amyloid-β-positive (n = 1242) versus negative (n = 536) participants. This preclinical AD network was similar to a previously published atrophy network associated with AD dementia (r = 0.8284, p = 0.016). In leave-one-out cross-validation, atrophy patterns connected to the preclinical AD network were associated with lower cognitive performance (p = 0.0018), greater subjective cognitive decline (p < 0.001), and amyloid-β levels (p < 0.001). Atrophy in preclinical AD maps to a network similar to AD dementia that is associated with amyloid-β and cognition, demonstrating an atrophy-related network across the continuum of AD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Reisa Sperling
- Harvard Aging Brain Study, Department of Neurology, Massachusetts General Hospital, Boston, MA 02114;Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's
| | - Nancy Donovan
- Brigham and Women's Hospital, Massachusetts General Hospital
| | - Shan Siddiqi
- Brigham and Women's Hospital, Massachusetts General Hospital
| |
Collapse
|
34
|
Vinay R, Probst J, Huynh P, Schlögl M, Kowatsch T, Nißen M. Top-funded digital health companies offering lifestyle interventions for dementia prevention: Company overview and evidence analysis. PLoS One 2025; 20:e0323390. [PMID: 40323948 PMCID: PMC12052105 DOI: 10.1371/journal.pone.0323390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 04/08/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND AND OBJECTIVE Dementia prevention has been recognized as a top priority by public health authorities due to the lack of a reversible cure. In this regard, digital dementia-preventive lifestyle services (DDLS) emerge as potentially pivotal services, aiming to address modifiable risk factors on a large scale. This study aims to identify the top-funded companies offering DDLS and evaluate their clinical evidence to gain insights into the international service landscape. METHODS A systematic screening of two financial databases (Pitchbook and Crunchbase) was conducted. Corresponding published clinical evidence was collected through a systematic literature review and analyzed regarding study purpose, results, quality of results, and level of clinical evidence. FINDINGS The ten top-funded companies offering DDLS received a total funding of EUR 128.52 million, of which three companies collected more than 75%. Clinical evidence was limited due to only nine eligible publications, small clinical subject groups, the absence of longitudinal study designs, and no direct evidence of dementia prevention. CONCLUSION Our study shows that the level of funding received by companies does not reflect the clinical effectiveness of DDLS. The study serves as an initial step toward understanding how DDLS are currently evaluated in today's market and highlights the need for a more rigorous evaluation of DDLS effectiveness.
Collapse
Affiliation(s)
- Rasita Vinay
- Institute of Biomedical Ethics and History of Medicine, University of Zurich, Zurich, Switzerland
- School of Medicine, University of St. Gallen, St. Gallen, Switzerland
- Centre for Digital Health Interventions, Department of Management, Technology and Economics, ETH Zurich, Zurich, Switzerland
| | - Jonas Probst
- School of Medicine, University of St. Gallen, St. Gallen, Switzerland
| | - Panitda Huynh
- School of Medicine, University of St. Gallen, St. Gallen, Switzerland
| | | | - Tobias Kowatsch
- School of Medicine, University of St. Gallen, St. Gallen, Switzerland
- Centre for Digital Health Interventions, Department of Management, Technology and Economics, ETH Zurich, Zurich, Switzerland
- Institute for Implementation Science in Health Care, University of Zurich, Zurich, Switzerland
| | - Marcia Nißen
- School of Medicine, University of St. Gallen, St. Gallen, Switzerland
- Centre for Digital Health Interventions, Department of Management, Technology and Economics, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
35
|
Chun MY, Park YH, Kim HJ, Na DL, Kim JP, Seo SW, Jang H. Distinct Characteristics of Suspected Non-Alzheimer Pathophysiology in Relation to Cognitive Status and Cerebrovascular Burden. Clin Nucl Med 2025; 50:368-380. [PMID: 40025666 PMCID: PMC11969373 DOI: 10.1097/rlu.0000000000005793] [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: 11/18/2024] [Accepted: 01/23/2025] [Indexed: 03/04/2025]
Abstract
PURPOSE OF THE REPORT This study investigated the prevalence and clinical characteristics of suspected non-Alzheimer disease pathophysiology (SNAP) across varying cognitive statuses and cerebral small vessel disease (CSVD) burden. PATIENTS AND METHODS We included 1992 participants with cognitive status categorized as cognitively unimpaired, mild cognitive impairment, or dementia. β-amyloid (Aβ, A) positivity was assessed by Aβ PET, and neurodegeneration (N) positivity was determined through hippocampal volume. Participants were further divided by the presence or absence of severe CSVD. The clinical and imaging characteristics of A-N+ (SNAP) group were compared with those of the A-N- and A+N+ groups. RESULTS SNAP participants were older and had more vascular risk factors compared with A-N- and A+N+ in the CSVD(-) cohort. SNAP and A+N+ showed similar cortical thinning. At the dementia stage, SNAP had a cognitive trajectory similar to A+N+ in the CSVD(-) cohort. However, SNAP exhibited less cognitive decline than A+N+ in the CSVD(+) cohort. CONCLUSIONS SNAP is characterized by distinct clinical and imaging characteristics; however, it does not necessarily indicate a benign prognosis, particularly at the dementia stage. These findings highlight the need to assess SNAP in relation to the cognitive stage and CSVD presence to better understand its progression and guide interventions.
Collapse
Affiliation(s)
- Min Young Chun
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine
- Department of Neurology, Yonsei University College of Medicine
| | - Yu Hyun Park
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Neuroscience Center, Samsung Medical Center
- Alzheimer’s Disease Convergence Research Center, Samsung Medical Center
| | - Hee Jin Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Alzheimer’s Disease Convergence Research Center, Samsung Medical Center
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University
- Department of Digital Health, SAIHST, Sungkyunkwan University
| | - Duk L. Na
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Alzheimer’s Disease Convergence Research Center, Samsung Medical Center
| | - Jun Pyo Kim
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Alzheimer’s Disease Convergence Research Center, Samsung Medical Center
| | - Sang Won Seo
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Alzheimer’s Disease Convergence Research Center, Samsung Medical Center
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University
- Department of Digital Health, SAIHST, Sungkyunkwan University
- Department of Clinical Research Design & Evaluation, SAIHST, Sungkyunkwan University, Gangnam-gu
| | - Hyemin Jang
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Jongno-gu, Seoul, South Korea
| |
Collapse
|
36
|
Cretin B. Epileptic variant in the spectrum of Alzheimer's disease - practical implications. Seizure 2025; 128:133-139. [PMID: 39343706 DOI: 10.1016/j.seizure.2024.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 09/11/2024] [Accepted: 09/20/2024] [Indexed: 10/01/2024] Open
Abstract
Alzheimer's disease (AD) is known to be associated with an increased risk of epilepsy, which is not exclusively related to the late stage of the disease - when a major cognitive impairment is observed, previously known as the dementia stage - but also to its prodromal stage (mild cognitive impairment). Moreover, published case reports and cohorts have shown that epilepsy may occur even earlier, at the preclinical stage of AD: Epileptic seizures may therefore be the sole objective manifestation of the disease. Such a situation is called the epileptic variant of AD (evAD). EvAD is one of the etiologies of late-onset epilepsy, which means that it carries a risk of later progression to dementia and that it can only be diagnosed by assessing amyloid and tau biomarkers. However, evAD is a window of therapeutic opportunity that is probably optimal for preventing, through antiseizure medication treatment, the accelerated cognitive decline associated with AD-related brain hyperexcitability (manifested by seizures or interictal epileptiform activities).
Collapse
Affiliation(s)
- Benjamin Cretin
- Centre Mémoire, de Ressources et de Recherche de Strasbourg, France; Unité de Neuropsychologie, Service de Neurologie des Hôpitaux Universitaires de Strasbourg, Strasbourg, France; University of Strasbourg and CNRS, ICube laboratory UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), team IMIS/Neurocrypto Strasbourg, France; Centre de Compétences des démences rares des Hôpitaux Universitaires de Strasbourg, France.
| |
Collapse
|
37
|
Kiene F, Hildebrandt H, Roheger M. Towards characterizing subjective cognitive decline in older adults. J Alzheimers Dis 2025; 105:609-621. [PMID: 40241516 DOI: 10.1177/13872877251330149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
BackgroundSubjective cognitive decline (SCD), where older adults perceive a persistent decline of cognitive abilities without showing an objective cognitive impairment, may represent a preclinical stage of Alzheimer's disease (AD) in some individuals.ObjectiveThe complex characteristics of SCD cannot only be revealed by existing self-report questionnaires. Rather, it is necessary to involve individuals affected in the research process with methods like focus group discussions (FGDs).MethodsStudy conduction took place in three steps: telephone interview, neuropsychological assessment and questionnaires, four FGDs with 16 older adults (11 female, 5 male) affected by SCD. FGDs were analyzed with qualitative content analysis using an inductive - deductive code system.ResultsAlthough the neuropsychological assessments did not indicate a cognitive impairment, participants reported a decline for all cognitive domains within the FGDs, especially for the memory- and speech domain, with declining word-finding abilities as the most salient symptom. Participants reported strong concerns related to SCD and difficulties in social participation.ConclusionsSCD seems to go beyond age-related cognitive changes, but as individuals do not show an objective cognitive impairment (yet), their symptoms are often not taken seriously enough. The FGDs revealed information that questionnaires or neuropsychological tests do not capture. The gained insight into SCD symptoms, related coping strategies and concerns is important to be able to develop measures for identifying individuals at risk for a transition to AD and to develop intervention measures that aim at delaying a further decline and increasing the quality of life of individuals affected.
Collapse
Affiliation(s)
- Franziska Kiene
- Ambulatory Assessment in Psychology, Department of Psychology, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| | - Helmut Hildebrandt
- Department of Psychology, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
- Department of Neurology, Hospital Bremen-Ost, Bremen, Germany
| | - Mandy Roheger
- Ambulatory Assessment in Psychology, Department of Psychology, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| |
Collapse
|
38
|
Maheshwari S, Singh A, Verma A. Biomarkers in Alzheimer's disease: new frontiers with olfactory models. Inflammopharmacology 2025; 33:2623-2640. [PMID: 40312605 DOI: 10.1007/s10787-025-01705-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 02/14/2025] [Indexed: 05/03/2025]
Abstract
Alzheimer's disease (AD), the leading cause of dementia worldwide, presents a significant diagnostic challenge, as clinical diagnoses are often made at advanced stages when neurodegenerative damage is already extensive. The study of biomarkers is necessary for improving identification, prognosis, and disease monitoring. Current research has primarily focused on cerebrospinal fluid and imaging biomarkers, including amyloid-β (Aβ1-42), phosphorylated tau, and total tau. However, these methods are invasive, expensive, and not widely accessible. Emerging approaches aim to identify novel, cost-effective, and minimally invasive biomarkers, particularly from blood-based and other peripheral sources. This review explores the role of olfactory neuronal precursors (ONPs) derived from the olfactory neuroepithelium (ONE) as a promising and innovative model for biomarker discovery in AD. ONPs can be non-invasively obtained directly from patients, offering a unique resource to study AD-related pathophysiological mechanisms. These neuronal lineage cells exhibit characteristics that make them a reliable surrogate model for central nervous system studies, enabling the evaluation of established biomarkers and facilitating the identification of novel candidates. Additionally, we discuss the potential of ONPs to enhance clinical practice through their accessibility and suitability for high-throughput biomarker analysis. By integrating the study of ONPs with existing biomarker research, this review highlights new frontiers in the quest to refine diagnostic tools and advance our understanding of Alzheimer's disease, paving the way for innovative strategies in early detection and personalized management.
Collapse
Affiliation(s)
- Shubhrat Maheshwari
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, 211007, India
| | - Aditya Singh
- Department of Pharmacy, Faculty of Pharmacy, Integral University, Lucknow, 226026, Uttar Pradesh, India
| | - Amita Verma
- Bioorganic and Medicinal Chemistry Research Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, 211007, India.
| |
Collapse
|
39
|
Laakko E, Oura P. Neurodegenerative disease findings in Finnish neuropathologically examined medico-legal autopsy cases with acute head injuries. J Forensic Leg Med 2025; 112:102866. [PMID: 40203702 DOI: 10.1016/j.jflm.2025.102866] [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: 09/15/2024] [Revised: 03/24/2025] [Accepted: 03/30/2025] [Indexed: 04/11/2025]
Abstract
Neurodegenerative diseases present diagnostic challenges due to their diverse clinical presentations, which emphasizes the importance of postmortem neuropathological examination. As neurodegenerative diseases become more common with age, they contribute to a higher risk of falls, accidents and head injuries. Our aims were to, first, report the prevalence of neurodegenerative disease findings in Finnish neuropathologically examined medico-legal autopsy cases with acute head injuries and, second, report the prevalence, injury circumstances, and types of specific brain injuries in the most common disease entities. We analyzed cases from the Helsinki office of the Forensic Medicine Unit, Finnish Institute for Health and Welfare, over the period 2016-2022. Cases were included if they had a suspected or confirmed head injury, and underwent a full neuropathological examination with neurodegenerative immunohistochemistry. Data on background characteristics, neurodegenerative immunostains and diagnoses, and head injuries were collected from cause-of-death investigation documents and neuropathology reports. Prevalences of neurodegenerative diagnoses were calculated, and head injury findings were presented for the most common disease entities. During the study period, there were a total of 128 neuropathologically examined cases with suspected or confirmed acute head injuries. Neurodegenerative immunohistochemistry was performed in 57 of them, and a neurodegenerative diagnosis was established in 47 cases. The most prevalent diagnoses were Alzheimer's disease neuropathologic change (ADNPC) (n = 34), cerebrovascular disease (CVD) (n = 34) and Lewy body disease (LBD) (n = 9). Old head injuries were common among cases with any neurodegenerative diagnosis (36.2 %). Of specific findings, subdural haemorrhage and hypoxic-ischaemic neuronal injury had the highest absolute prevalence in cases with ADNPC, although there were no major differences between ADNPC, CVD and LBD. In conclusion, the most prevalent neurodegenerative disease entities in this dataset were ADNPC, CVD and LBD. A history of head injuries was common regardless of the neurodegenerative entity.
Collapse
Affiliation(s)
- Essi Laakko
- Department of Forensic Medicine, University of Helsinki, P.O. Box 21, FI-00014, Helsinki, Finland
| | - Petteri Oura
- Department of Forensic Medicine, University of Helsinki, P.O. Box 21, FI-00014, Helsinki, Finland; Forensic Medicine Unit, Finnish Institute for Health and Welfare, P.O. Box 30, FI-00271, Helsinki, Finland.
| |
Collapse
|
40
|
Cohen AD, Villemagne VL. A brief history of Aβ imaging. Alzheimers Dement 2025; 21:e70291. [PMID: 40407091 PMCID: PMC12100503 DOI: 10.1002/alz.70291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/26/2025]
Abstract
β-Amyloid (Αβ) imaging revolutionized the in vivo assessment of Alzheimer's disease (AD) Αβ pathology and its changes over time, increasing our insights into Aβ deposition in the brain by providing highly accurate, reliable, and reproducible quantitative statements of regional and global Aβ burden in the brain, proving essential for the differential diagnosis, staging, and evaluation of disease-specific anti-Αβ therapeutic approaches. Longitudinal observations, coupled with different disease-specific biomarkers to assess potential downstream effects of Aβ, have confirmed that Αβ deposition in the brain starts decades before the onset of symptoms. Aβ imaging studies continue to refine our understanding of the role of Αβ deposition in AD, and its relation to other imaging and fluid biomarkers. HIGHLIGHTS: Αβ imaging revolutionized the in vivo assessment of Alzheimer's disease Αβ pathology. Αβ imaging has increased our insights into Aβ deposition in the brain by providing highly accurate, reliable, and reproducible quantitative statements of regional and global Αβ burden in the brain. Αβ imaging is essential for the differential diagnosis, staging, and evaluation of disease-specific anti-Αβ therapeutic approaches. Αβ imaging studies continue to refine our understanding of the role of Αβ deposition in Alzheimer's disease, and its relation to other imaging and fluid biomarkers.
Collapse
Affiliation(s)
- Ann D. Cohen
- Department of PsychiatryThe University of PittsburghPittsburghPennsylvaniaUSA
| | | |
Collapse
|
41
|
Wei M, Wang L, Yu X, Hu W, Wang M, Zhang Q, Guo T, Zhong J, Li C, Jiang J, Han Y. Differences in Glucose Metabolism Between Single Memory Domain and Multidomain Subjective Cognitive Decline: A Longitudinal Study From SILCODE. CNS Neurosci Ther 2025; 31:e70264. [PMID: 40426276 PMCID: PMC12116337 DOI: 10.1111/cns.70264] [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: 04/23/2024] [Revised: 12/22/2024] [Accepted: 01/30/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Glucose metabolism and plasma biomarkers have emerged as important early markers in Alzheimer's disease. Different subtypes (single memory domain, multidomain) of subjective cognitive decline (SCD) may represent distinct stages of disease progression, but the differences in glucose metabolism remain unclear. This study focused on exploring the differences in glucose metabolism between different SCD subtypes and the correlation with plasma biomarkers based on 18F-FDG PET. METHODS In this study, thirty-three normal controls (NCs), thirty-five individuals with single memory domain SCD (sd-SCD), thirty-nine individuals with multidomain SCD (md-SCD), and twenty-one cognitively impaired (CI) individuals were involved. We investigated the standardized uptake value ratio (SUVR) and voxel differences between the sd-SCD and md-SCD groups followed by FDR and GRF corrections, with an average follow-up time of 44.98 ± 16.49 months. Correlation analyses were employed to assess relationships between FDG-PET SUVR and neuropsychological scales as well as plasma biomarkers. Finally, Kaplan-Meier survival analysis was used to investigate the risk of cognitive decline conversion among SCD subgroups. RESULTS After controlling for the effects of covariates, the following brain regions showed voxel differences and lower SUVR in md-SCD groups, including right anterior cingulate and paracingulate gyri (ACG.R, p = 0.003), left anterior cingulate and paracingulate gyri (ACG.L, p = 0.003), right middle temporal gyrus (MTG.R, p = 0.004), and right inferior temporal gyrus (ITG.R, p = 0.001), compared to the sd-SCD group. SUVR of ACG.R was correlated with plasma Aβ42/40 (r = 0.435, p = 0.006) and AVLT-N7 score (r = 0.347, p = 0.031) in the md-SCD group while none of the correlations existed in the sd-SCD group. SUVR of MTG.R was also correlated with the AVLT-N7 score (r = 0.246, p = 0.035) across SCD individuals. The SCD individuals with positive plasma Aβ42/40, p-tau181, and glucose metabolism in above four regions, or those in the md-SCD group showed an elevated risk of cognitive conversion in comparison to the controls. CONCLUSIONS Differences in glucose metabolism could be observed between the md-SCD and sd-SCD groups. SCD participants in the md-SCD group, or those with positive biomarkers, might represent a higher risk of cognitive decline conversion.
Collapse
Affiliation(s)
- Min Wei
- Department of NeurologyXuanwu Hospital of Capital Medical UniversityBeijingChina
| | - Luyao Wang
- Institute of Biomedical Engineering, School of Life SciencesShanghai UniversityShanghaiChina
| | - Xianfeng Yu
- Department of NeurologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Wenjing Hu
- Institute of Biomedical Engineering, School of Life SciencesShanghai UniversityShanghaiChina
| | - Min Wang
- Institute of Biomedical Engineering, School of Life SciencesShanghai UniversityShanghaiChina
| | - Qi Zhang
- Institute of Biomedical Engineering, School of Life SciencesShanghai UniversityShanghaiChina
| | - Tengfei Guo
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
| | - Jiayi Zhong
- Institute of Biomedical Engineering, School of Life SciencesShanghai UniversityShanghaiChina
| | - Chenyang Li
- Institute of Biomedical Engineering, School of Life SciencesShanghai UniversityShanghaiChina
| | - Jiehui Jiang
- Institute of Biomedical Engineering, School of Life SciencesShanghai UniversityShanghaiChina
| | - Ying Han
- Department of NeurologyXuanwu Hospital of Capital Medical UniversityBeijingChina
- Institute of Biomedical EngineeringShenzhen Bay LaboratoryShenzhenChina
- State Key Laboratory of Digital Medical Engineering, Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical EngineeringHainan UniversitySanyaChina
- National Clinical Research Center for Geriatric DiseasesBeijingChina
- The Central Hospital of KaramayXinjiangChina
| |
Collapse
|
42
|
Goswami S, Hartz SM, Oliver A, Jackson S, Ogungbenle T, Evans A, Linnenbringer E, Moulder KL, Morris JC, Mozersky J. Research Participant Interest in Learning Results of Biomarker Tests for Alzheimer Disease. JAMA Netw Open 2025; 8:e252919. [PMID: 40327345 PMCID: PMC12056564 DOI: 10.1001/jamanetworkopen.2025.2919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 01/29/2025] [Indexed: 05/07/2025] Open
Abstract
Importance While available evidence suggests there are not major psychosocial harms of return of research results, there are limited data on uptake of Alzheimer disease (AD) research results or reasons for declining to learn results among participants in AD research studies. Objectives To quantitatively and qualitatively evaluate who declines to learn individual AD biomarker research results and what factors are associated with the decision. Design, Setting, and Participants This observational cohort study was conducted between November 1, 2020, and April 15, 2024, among participants aged 65 years or older with unimpaired cognition and available biomarker data (apolipoprotein E genotype and either imaging [amyloid positron emission tomography and magnetic resonance imaging] or plasma amyloid level) enrolled in a longitudinal cohort of cognitive aging at the Knight Alzheimer Disease Research Center. Exposure Participants with no prior option to receive research results were offered the option to learn these results. Main Outcome and Measures The primary outcome was the decision to receive AD research biomarker results. Associations of this decision with demographic factors including self-identified race, parental history of AD, age, gender, and type of biomarker result offered (imaging or plasma) were assessed using χ2 tests and semiparametric log-binomial regression. Semistructured qualitative interviews with a subset of participants who declined receiving research results explored reasons for declining. Results Of the 274 participants who were offered their research results (mean [SD] age, 75.9 [5.8] years; 158 women [58%]; 35 Black [13%]; and 239 White [87%]), 110 (40%) declined. Black participants were more likely to decline than White participants (adjusted risk ratio, 1.89; 95% CI, 1.43-2.50). Participants with a known parental history of AD dementia were more likely to decline than those without (adjusted risk ratio, 1.49; 95% CI, 1.12-1.98). Qualitative interviews found the following reasons for declining: knowing would be a burden, negative experiences and perceptions of AD dementia, feeling good about memory currently, familial burden, already being prepared, and the uncertainty of results. Conclusions and Relevance In this study of participants enrolled in a longitudinal aging study offered their research results, those with a parental history of AD dementia and Black participants were significantly more likely to decline. Qualitative interviews suggest that a family history of AD may create negative experiences and perceptions of the disease, which may influence the decision to learn results. Further research is needed to better understand racial differences in uptake and ensure that the choice to receive research results reflects individual preferences and wishes.
Collapse
Affiliation(s)
- Spondita Goswami
- Bioethics Research Center, Washington University in St Louis, St Louis, Missouri
- Knight Alzheimer Disease Research Center, Washington University in St Louis, St Louis, Missouri
| | - Sarah M. Hartz
- Knight Alzheimer Disease Research Center, Washington University in St Louis, St Louis, Missouri
- Department of Psychiatry, Washington University in St Louis, St Louis, Missouri
| | - Amy Oliver
- Knight Alzheimer Disease Research Center, Washington University in St Louis, St Louis, Missouri
- Department of Psychiatry, Washington University in St Louis, St Louis, Missouri
| | - Sacha Jackson
- Knight Alzheimer Disease Research Center, Washington University in St Louis, St Louis, Missouri
- Department of Psychiatry, Washington University in St Louis, St Louis, Missouri
| | - Tomi Ogungbenle
- Department of Health Care Ethics, Saint Louis University, St Louis, Missouri
| | - Alissa Evans
- Knight Alzheimer Disease Research Center, Washington University in St Louis, St Louis, Missouri
- Department of Psychiatry, Washington University in St Louis, St Louis, Missouri
| | - Erin Linnenbringer
- Division of Public Health Sciences, Washington University in St Louis, St Louis, Missouri
| | - Krista L. Moulder
- Knight Alzheimer Disease Research Center, Washington University in St Louis, St Louis, Missouri
| | - John C. Morris
- Knight Alzheimer Disease Research Center, Washington University in St Louis, St Louis, Missouri
| | - Jessica Mozersky
- Bioethics Research Center, Washington University in St Louis, St Louis, Missouri
- Knight Alzheimer Disease Research Center, Washington University in St Louis, St Louis, Missouri
| |
Collapse
|
43
|
Beltrami D, Barletta-Rodolfi C, Bertini F, Braglia L, Calzà L, Corbo M, Gasparini F, Marti A, Montesi D, Pisano M, Rusconi ML, Sozzi M, Tonon C, Ghidoni E. Normative data for COGITAB: An Italian tablet-based test battery conceived for the preclinical phase of Alzheimer's disease. APPLIED NEUROPSYCHOLOGY. ADULT 2025; 32:664-674. [PMID: 37289131 DOI: 10.1080/23279095.2023.2219797] [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: 06/09/2023]
Abstract
The number of people with dementia is increasing worldwide. Two main approaches have been adopted to identify subjects with Alzheimer's disease (AD): the neuropsychological evaluation and the identification of biomarkers of AD. The first method is less invasive and easier to perform. This study assesses the psychometric properties of COGITAB, a novel web application d esigned to be sensitive to the subtle cognitive changes distinctive of the early Mild Cognitive Impairment (MCI) and the preclinical phase of AD. We enrolled 518 healthy controls, classified according to several risk factors and the presence of a family history of dementia. The participants were given COGITAB after a neuropsychological screening. The COGITAB Total Score (TS) was significantly affected by age and years of education. Acquired risk factors and family history of dementia significantly impacted only the COGITAB total execution time (TET), not the TS. This study provides normative data for a newly developed web application. Control subjects with acquired risk factors performed slower, giving an important role to the TET recording. Further studies should examine the ability of this new technology to discriminate between healthy subjects and subjects with initial cognitive decline, even when not detected by standard neuropsychological assessments.
Collapse
Affiliation(s)
- Daniela Beltrami
- Clinical Neuropsychology, Cognitive Disorders and Dyslexia Unit, Neurology, Department of Neuro-Motor Diseases, AUSL IRCCS, Reggio Emilia, Italy
| | - Caterina Barletta-Rodolfi
- Clinical Neuropsychology, Cognitive Disorders and Dyslexia Unit, Neurology, Department of Neuro-Motor Diseases, AUSL IRCCS, Reggio Emilia, Italy
| | - Flavio Bertini
- Department of Mathematical, Physical and Computer Sciences, University of Parma, Parma, Italy
| | - Luca Braglia
- Research and Statistics Infrastructure, AUSL - IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Laura Calzà
- Interdepartmental Centre for Industrial Research in Health Sciences and Technologies, University of Bologna, Bologna, Italy
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Massimo Corbo
- Department of Neurorehabilitation Sciences, Casa Cura Igea (CCI), Milano, Italy
| | - Federico Gasparini
- Clinical Neuropsychology, Cognitive Disorders and Dyslexia Unit, Neurology, Department of Neuro-Motor Diseases, AUSL IRCCS, Reggio Emilia, Italy
| | - Alessandro Marti
- Clinical Neuropsychology, Cognitive Disorders and Dyslexia Unit, Neurology, Department of Neuro-Motor Diseases, AUSL IRCCS, Reggio Emilia, Italy
| | - Danilo Montesi
- SmartData Research Group, Department of Computer Science and Engineering, University of Bologna, Bologna, Italy
| | - Marta Pisano
- Clinical Neuropsychology, Cognitive Disorders and Dyslexia Unit, Neurology, Department of Neuro-Motor Diseases, AUSL IRCCS, Reggio Emilia, Italy
| | - Maria Luisa Rusconi
- Department of Human and Social Sciences, University of Bergamo, Bergamo, Italy
| | - Matteo Sozzi
- Department of Neuroscience, Neurology. ASST "A. Manzoni", Lecco, Italy
| | - Cecilia Tonon
- Clinical Neuropsychology, Cognitive Disorders and Dyslexia Unit, Neurology, Department of Neuro-Motor Diseases, AUSL IRCCS, Reggio Emilia, Italy
| | - Enrico Ghidoni
- Clinical Neuropsychology, Cognitive Disorders and Dyslexia Unit, Neurology, Department of Neuro-Motor Diseases, AUSL IRCCS, Reggio Emilia, Italy
| |
Collapse
|
44
|
Akinci M, Aguilar‐Domínguez P, Palpatzis E, Shekari M, García‐Prat M, Deulofeu C, Fauria K, García‐Aymerich J, Gispert JD, Suárez‐Calvet M, Grau‐Rivera O, Sánchez‐Benavides G, Arenaza‐Urquijo EM, for the ALFA study. Physical activity changes during midlife link to brain integrity and amyloid burden. Alzheimers Dement 2025; 21:e70007. [PMID: 40304268 PMCID: PMC12042120 DOI: 10.1002/alz.70007] [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: 11/15/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 05/02/2025]
Abstract
INTRODUCTION Evidence suggests that midlife physical activity may reduce Alzheimer's disease (AD) risk. In at-risk individuals, we investigated midlife physical activity changes in relation to AD-related pathologies. METHODS We included 337 cognitively unimpaired adults with baseline and follow-up physical activity evaluations within 4.07 ± 0.84 years. We performed multiple regressions considering follow-up amyloid-PET burden and MRI-based medial temporal lobe cortical thickness as outcomes. Independent variables encompassed changes in adherence to World Health Organization (WHO)-recommended physical activity levels, activity amounts, and sedentary behavior (no activity reported). RESULTS Remaining sedentary was associated with lower cortical thickness compared to doing limited physical activity, maintaining adherence, or becoming adherent to WHO recommendations. Becoming adherent to recommendations was linked to lower amyloid burden compared to becoming non-adherent. Increased activity amounts showed a dose-dependent association with lower amyloid burden. DISCUSSION Increasing physical activity and new adherence to WHO recommendations could be key objectives for preventive strategies during midlife. CLINICAL TRIAL REGISTRATION INFORMATION Registered at Clinicaltrials.gov (identifier: NCT02485730). HIGHLIGHTS Boosting physical activity in midlife may have beneficial effects in preclinical AD. Physical activity increases relate to lower Aβ burden in a dose-dependent manner. Remaining sedentary links to lower cortical thickness in AD-vulnerable structures. New adherence to WHO-recommended physical activity levels may enhance brain health.
Collapse
Affiliation(s)
- Muge Akinci
- Barcelona Institute for Global Health (ISGlobal)BarcelonaSpain
- Barcelonaβeta Brain Research Center ‐ Pasqual Maragall FoundationBarcelonaSpain
- Universitat Pompeu Fabra (UPF)BarcelonaSpain
- CIBER Epidemiología y Salud Pública (CIBERESP)MadridSpain
| | - Pablo Aguilar‐Domínguez
- Barcelona Institute for Global Health (ISGlobal)BarcelonaSpain
- Universitat Pompeu Fabra (UPF)BarcelonaSpain
- CIBER Epidemiología y Salud Pública (CIBERESP)MadridSpain
- Sant Pau Memory Unit, Department of NeurologyHospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIBSant Pau)BarcelonaSpain
| | - Eleni Palpatzis
- Barcelona Institute for Global Health (ISGlobal)BarcelonaSpain
- Barcelonaβeta Brain Research Center ‐ Pasqual Maragall FoundationBarcelonaSpain
- Universitat Pompeu Fabra (UPF)BarcelonaSpain
- CIBER Epidemiología y Salud Pública (CIBERESP)MadridSpain
| | - Mahnaz Shekari
- Barcelonaβeta Brain Research Center ‐ Pasqual Maragall FoundationBarcelonaSpain
- Universitat Pompeu Fabra (UPF)BarcelonaSpain
- Hospital del Mar Medical Research InstituteBarcelonaSpain
| | - Marina García‐Prat
- Barcelonaβeta Brain Research Center ‐ Pasqual Maragall FoundationBarcelonaSpain
| | - Carme Deulofeu
- Barcelonaβeta Brain Research Center ‐ Pasqual Maragall FoundationBarcelonaSpain
| | - Karine Fauria
- Barcelonaβeta Brain Research Center ‐ Pasqual Maragall FoundationBarcelonaSpain
| | - Judith García‐Aymerich
- Barcelona Institute for Global Health (ISGlobal)BarcelonaSpain
- Universitat Pompeu Fabra (UPF)BarcelonaSpain
- CIBER Epidemiología y Salud Pública (CIBERESP)MadridSpain
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center ‐ Pasqual Maragall FoundationBarcelonaSpain
- Universitat Pompeu Fabra (UPF)BarcelonaSpain
- Hospital del Mar Medical Research InstituteBarcelonaSpain
- Centro de Investigación Biomédica en Red de BioingenieríaBiomateriales y Nanomedicina (CIBER‐BBN)MadridSpain
| | - Marc Suárez‐Calvet
- Barcelonaβeta Brain Research Center ‐ Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Medical Research InstituteBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
- Servei de NeurologiaHospital del MarBarcelonaSpain
| | - Oriol Grau‐Rivera
- Barcelonaβeta Brain Research Center ‐ Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Medical Research InstituteBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
- Servei de NeurologiaHospital del MarBarcelonaSpain
| | - Gonzalo Sánchez‐Benavides
- Barcelonaβeta Brain Research Center ‐ Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Medical Research InstituteBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
| | - Eider M. Arenaza‐Urquijo
- Barcelona Institute for Global Health (ISGlobal)BarcelonaSpain
- Barcelonaβeta Brain Research Center ‐ Pasqual Maragall FoundationBarcelonaSpain
- Universitat Pompeu Fabra (UPF)BarcelonaSpain
- CIBER Epidemiología y Salud Pública (CIBERESP)MadridSpain
- Hospital del Mar Medical Research InstituteBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
| | | |
Collapse
|
45
|
Ostlund SB, Chen G, Kosheleff A, Lueptow LM, Zhuravka I, Frautschy SA, Lam HA, Maidment NT. Early emergence of motivational and hedonic feeding deficits in the TgF344-AD rat model of Alzheimer's disease. Front Aging Neurosci 2025; 17:1572956. [PMID: 40357232 PMCID: PMC12066702 DOI: 10.3389/fnagi.2025.1572956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/08/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction Alzheimer's disease (AD) is characterized by progressive cognitive decline and has a long prodromal phase during which subclinical cognitive deficits and neuropsychiatric symptoms may begin to emerge. Apathy, defined as a lack of motivation or volition, is increasingly recognized as a core feature and a potentially early marker of AD. Despite its significance, apathy-like behavior has been underexplored in transgenic models of AD. Methods We performed a longitudinal analysis of apathy-like behavior using the well-established TgF344-AD rat model. We compared male and female TgF344-AD and wildtype rats on hedonic (palatable food intake) and motivational (progressive ratio) assays during early (3-4 months), intermediate (6-7 months), and later (9-10 months) stages of adulthood. Results We found that female TgF344-AD rats exhibited early and persistent deficits in motivational and hedonic feeding, emerging at 3-4 months and 6-7 months, respectively. During a battery of cognitive tests conducted after 12-14 months of age, TgF344-AD rats were impaired in spatial working memory but also showed wide-ranging deficits in exploratory behavior, which may also be indicative of an apathy-like loss of investigatory drive. Conclusion Our findings highlight the TgF344-AD rat as a valuable model for studying early apathy-like behavior in AD and underscore the need to consider sex differences in AD research to better understand the prodromal phase of this disease.
Collapse
Affiliation(s)
- Sean B. Ostlund
- Department of Anesthesiology and Perioperative Care, Irvine School of Medicine, Irvine Center for Addiction Neuroscience (ICAN), University of California, Irvine, Irvine, CA, United States
| | - Grace Chen
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Alisa Kosheleff
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Lindsay M Lueptow
- Behavioral Testing Core, Department of Psychology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Irina Zhuravka
- Behavioral Testing Core, Department of Psychology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Sally A. Frautschy
- Departments of Neurology and Medicine, Geriatric Research Education and Clinical Center, Veterans Greater Los Angeles HealthCare System, University of California, Los Angeles, Los Angeles, CA, United States
| | - Hoa A. Lam
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Nigel T. Maidment
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
46
|
Colín-Martínez E, Arias C. Involvement of the VGF/BDNF axis in the neuropathology of Alzheimer's disease and its potential role in diagnosis and treatment. Rev Neurosci 2025; 36:267-278. [PMID: 39566031 DOI: 10.1515/revneuro-2024-0110] [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: 08/13/2024] [Accepted: 09/26/2024] [Indexed: 11/22/2024]
Abstract
The brain is a highly plastic organ that continually receives and integrates signals to generate functional and structural changes and homeostatic adaptations throughout life. Alterations in some signaling pathways that mediate these responses can impact brain plasticity, accelerate brain aging and potentially lead to neurodegeneration. There is substantial evidence that two important signaling pathways activated by neurotrophins, nonacronymic (VGF) and brain-derived neurotrophic factor (BDNF), are involved in substantial functions stimulating neuronal growth, differentiation, and circuit establishment during development and neuronal maintenance and plasticity in the mature brain. In this review, we present evidence that these two pathways and their interactions are central players in cognitive performance and alterations in pathological aging, particularly in conditions such as Alzheimer's disease (AD). Finally, we suggest specific avenues for future research on the basis of recent findings suggesting these molecules are diagnostic biomarkers and putative therapeutic tools to prevent, delay or improve AD neuropathology.
Collapse
Affiliation(s)
- Elizabeth Colín-Martínez
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, México
| | - Clorinda Arias
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, México
| |
Collapse
|
47
|
Henzen NA, Abdulkadir A, Reinhardt J, Blatow M, Kressig RW, Krumm S. Automated segmentation for cortical thickness of the medial perirhinal cortex. Sci Rep 2025; 15:14903. [PMID: 40295570 PMCID: PMC12037834 DOI: 10.1038/s41598-025-98399-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 04/11/2025] [Indexed: 04/30/2025] Open
Abstract
Alzheimer's disease (AD) is characterized by a progressive spread of neurofibrillary tangles (NFT), beginning in the medial perirhinal cortex (mPRC), advancing to the entorhinal cortex (ERC), and subsequently involving the hippocampus, lateral perirhinal cortex (lPRC), and the rest of the brain. Given the close relationship between NFT accumulation and neuronal loss, the mPRC reflects a promising structural marker for early diagnosis of AD. However, only limited tools that automatically measure the cortical thickness of the mPRC are currently available. Utilizing the nnU-Net framework, we trained models on structural MRI of 126 adults, with manually segmented labels as ground truth. These models were then applied to an independent dataset of 103 adults (comprising patients with Alzheimer's dementia, amnestic mild cognitive impairment (aMCI), and healthy controls). High agreement was observed between manual and automated measurements of cortical thickness. Furthermore, we found significant atrophy in the Alzheimer's dementia group in the mPRC, ERC, and lPRC compared to healthy controls. Comparison of the aMCI group and healthy controls revealed significant differences in the ERC only. The results underscore the utility of our automated segmentation tool in advancing Alzheimer's research.
Collapse
Affiliation(s)
- Nicolas A Henzen
- University Department of Geriatric Medicine FELIX PLATTER, Burgfelderstrasse 101, 4055, Basel, Switzerland.
- Faculty of Psychology, University of Basel, Basel, Switzerland.
| | - Ahmed Abdulkadir
- Department of Clinical Neurosciences, Laboratory for Research in Neuroimaging LREN, Centre for Research in Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Center for Artificial Intelligence, Zürich University of Applied Sciences, Winterthur, Switzerland
- University Hospital of Old Age Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
| | - Julia Reinhardt
- Division of Diagnostic and Interventional Neuroradiology, Department of Radiology, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Cardiology and Cardiovascular Research Institute Basel (CRIB), University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Orthopedic Surgery and Traumatology, University Hospital of Basel, University of Basel, Basel, Switzerland
| | - Maria Blatow
- Section of Neuroradiology, Department of Radiology and Nuclear Medicine, Neurocenter, Cantonal Hospital Lucerne, University of Lucerne, Lucerne, Switzerland
| | - Reto W Kressig
- University Department of Geriatric Medicine FELIX PLATTER, Burgfelderstrasse 101, 4055, Basel, Switzerland
- Faculty of Medicine, University of Basel, Basel, Switzerland
| | - Sabine Krumm
- University Department of Geriatric Medicine FELIX PLATTER, Burgfelderstrasse 101, 4055, Basel, Switzerland
- Faculty of Medicine, University of Basel, Basel, Switzerland
| |
Collapse
|
48
|
Kisli M, Saçmacı H. Pattern-VEP findings in individuals with mild cognitive impairment. Behav Brain Res 2025; 484:115504. [PMID: 40023256 DOI: 10.1016/j.bbr.2025.115504] [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: 09/24/2024] [Revised: 02/13/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND Visual evoked potential (VEP) is a technique used to evaluate the electrical response of the brain to visual stimuli. This study aimed to examine neural transmission in the visual pathway by VEP test in individuals with mild cognitive impairment (MCI) and compare it with age-appropriate controls and also investigate for a correlation between VEP parameters and cognitive test domains. METHODS The groups consisted of 56 MCI and 50 healthy volunteers, aged 60-80 years, matched for age and education. Mini-Mental State Examination (MMSE) was applied to the participants for cognitive assessment. Patients were also subjected to other dementia screening tests and other treatable causes were excluded. In addition, groups were formed from those who completed the test. The pattern-reversal VEP method was used in this study. RESULTS The mean MMSE score in the MCI group was 21.42 ± 1.55 points. Our findings showed that individuals with MCI had a longer left P100 latency compared to controls (p = 0.027). In addition, right N75-P100 and right P100-N145 amplitudes of VEP parameters were found to be positively correlated with the recall test, which is one of the MMSE domains (p < 0.05, rho: 0.399,0.314). CONCLUSION The evidence provided by this study supports the possibility of using pattern VEP with clinical parameters to evaluate MCI patients. In addition, a positive correlation between interpeak amplitudes and the recall test highlights the importance of the VEP test in these patients.
Collapse
Affiliation(s)
- Mesude Kisli
- Department of Neurology, Sivas State Hospital, Sivas, Turkey.
| | - Hikmet Saçmacı
- Department of Neurology, Bozok University School of Medicine, Yozgat, Turkey.
| |
Collapse
|
49
|
Mao C, Mo Y, Jiang J, Fang S, Hu Z, Ke Z, Zhao H, Xu Y. Association between high plasma p-tau181 level and gait changes in patients with mild cognitive impairment. Sci Rep 2025; 15:14679. [PMID: 40287471 PMCID: PMC12033327 DOI: 10.1038/s41598-025-94472-6] [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: 08/06/2024] [Accepted: 03/13/2025] [Indexed: 04/29/2025] Open
Abstract
Previous studies on gait changes in mild cognitive impairment (MCI) are inconsistent. Alzheimer's disease (AD) plasma biomarkers, amyloid beta (Aβ) and phosphorylated-tau (p-tau), are relevant to gait disorders. This study explores gait changes in MCI and the relationship between gait performance and AD plasma biomarkers. 231 participants were recruited and stratified based on p-tau181 levels into: low p-tau181 with normal cognition (lT-NC), low p-tau181 with MCI (lT-MCI), and high p-tau181 with MCI (hT-MCI). The same cohort was subsequently stratified by Aβ42/Aβ40 levels into: high Aβ42/Aβ40 with normal cognition (hA-NC), high Aβ42/Aβ40 with MCI (hA-MCI), and low Aβ42/Aβ40 with MCI (lA-MCI). Demographic, cognitive and gait data were compared across groups. The hT-MCI and lA-MCI groups were older than the other groups. Significant differences in stride length were found between lT-NC and hT-MCI, lT-MCI and hT-MCI, but not between lT-NC and lT-MCI. Neuropsychological assessments revealed poorer performance in hT-MCI and lT-MCI groups relative to lT-NC, while global cognitive function was comparable between hT-MCI and lT-MCI groups. No such associations were observed between stride length and Aβ42/Aβ40 levels. Decreased stride length, which is generally considered to be indicative of poorer gait, was significantly associated with elevated p-tau181 levels and independent of global cognitive status. These findings highlight the potential of p-tau181 as a biomarker for tau-related motor dysfunction in MCI.
Collapse
Affiliation(s)
- Chenglu Mao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China.
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210000, China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210000, China.
- Nanjing Neurology Clinical Medical Center, Nanjing, 210000, China.
| | - Yuting Mo
- Department of Neurology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, 210000, China
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210000, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210000, China
- Nanjing Neurology Clinical Medical Center, Nanjing, 210000, China
| | - Jialiu Jiang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210000, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210000, China
- Nanjing Neurology Clinical Medical Center, Nanjing, 210000, China
| | - Shuang Fang
- Department of Neurology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, 210000, China
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210000, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210000, China
- Nanjing Neurology Clinical Medical Center, Nanjing, 210000, China
| | - Zheqi Hu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210000, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210000, China
- Nanjing Neurology Clinical Medical Center, Nanjing, 210000, China
| | - Zhihong Ke
- Department of Neurology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, 210000, China
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210000, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210000, China
- Nanjing Neurology Clinical Medical Center, Nanjing, 210000, China
| | - Hui Zhao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China.
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210000, China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210000, China.
- Nanjing Neurology Clinical Medical Center, Nanjing, 210000, China.
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China.
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210000, China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210000, China.
- Nanjing Neurology Clinical Medical Center, Nanjing, 210000, China.
| |
Collapse
|
50
|
Yeh TS, Blacker D, Willett WC. Dietary Factors and Cognitive Function: with a Focus on Subjective Cognitive Decline. Curr Nutr Rep 2025; 14:62. [PMID: 40285979 DOI: 10.1007/s13668-025-00638-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2025] [Indexed: 04/29/2025]
Abstract
PURPOSE OF THE REVIEW Age-related cognitive decline is an important global challenge. Substantial evidence suggests that diet may prevent or delay cognitive aging. This narrative review examines recent literature on how dietary factors influence cognitive function, with a focus on subjective cognitive decline (SCD). RECENT FINDINGS Higher intakes of flavonoids, carotenoids, and plant-based protein were associated with lower odds of SCD. Berries, citrus fruits and juices, carotenoid-rich and green leafy vegetables, and beans/legumes were among the foods with the strongest inverse associations with SCD. Healthy dietary patterns, such as the Mediterranean and MIND diet, may be beneficial for maintaining subjective cognitive function. Healthy choice of diet may play a role in lowering the risk of late-life SCD.
Collapse
Affiliation(s)
- Tian-Shin Yeh
- Department of Physical Medicine and Rehabilitation, School of Medicine, College of Medicine, Taipei Medical University, No.250, Wuxing St, Taipei, 11031, Taiwan.
- Department of Physical Medicine and Rehabilitation, Shuang Ho Hospital, Taipei Medical University, New Taipei, 23561, Taiwan.
- Department of Physical Medicine and Rehabilitation, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
- Department of Nutrition and Epidemiology, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA, USA.
- Nuffield Department of Population Health, Big Data Institute, University of Oxford, Oxford, UK.
| | - Deborah Blacker
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Walter C Willett
- Department of Nutrition and Epidemiology, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA, USA
| |
Collapse
|