1
|
Abu Raya M, Zeltzer E, Blazhenets G, Schonhaut DR, Allen IE, Carrillo MC, Gatsonis C, Hanna L, Hillner BE, Iaccarino L, March A, Mundada NS, Perez JM, Smith K, Yballa C, Siegel BA, Windon C, Whitmer RA, Kaitlin C, La Joie R, Rabinovici GD. Sex differences in amyloid PET in a large, real-world sample from the Imaging Dementia-Evidence for Amyloid Scanning (IDEAS) Study. Alzheimers Dement 2025; 21:e70304. [PMID: 40415175 DOI: 10.1002/alz.70304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 05/27/2025]
Abstract
INTRODUCTION We examined sex effects on amyloid positron emission tomography (PET) in a large cohort of patients evaluated for cognitive complaints in a "real-world" specialty setting. METHODS We analyzed 10,361 amyloid PET scans (51% females) from the Imaging Dementia-Evidence for Amyloid Scanning Study. Amyloid positivity was defined by either local visual read or central PET processing and quantification (≥ 24.4 Centiloids). Sex differences were examined using multilinear regression and logistic regression adjusted for age, comorbidities, and other demographic and clinical covariates. RESULTS Females had higher rates of positive amyloid PET visual reads (63% vs. 59%, P < 0.001) and higher Centiloids (CLs; median 48.7 vs. 36.8, p < 0.001). On logistic regression, females had higher odds ratios (ORs) for positive amyloid PET (visual read OR 1.20, 95% confidence interval [CI]: 1.11-1.31; CL threshold-based OR 1.37, 95% CI: 1.26-1.49; both p < 0.001). DISCUSSION Females with cognitive impairment showed higher amyloid PET positivity and greater amyloid burden. Further research is needed to explore mechanisms and treatment implications. HIGHLIGHTS Females exhibited higher rates of amyloid positron emission tomography (PET) positivity and higher amyloid burden than males. These sex effects were found in patients with both mild cognitive impairment (MCI) and dementia. Females also had higher rates of dementia and amnestic MCI, while males had higher rates of non-amnestic MCI and more cholinesterase inhibitor use.
Collapse
Affiliation(s)
- Maison Abu Raya
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
- Global Brain Health Institute, University of California San Francisco, San Francisco, California, USA
| | - Ehud Zeltzer
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Ganna Blazhenets
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Daniel R Schonhaut
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Isabel Elaine Allen
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
- Global Brain Health Institute, University of California San Francisco, San Francisco, California, USA
| | - Maria C Carrillo
- Medical & Scientific Relations Division, Alzheimer's Association, Chicago, Illinois, USA
| | | | - Lucy Hanna
- Center for Statistical Sciences, Brown University, Providence, Rhode Island, USA
| | - Bruce E Hillner
- Department of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Leonardo Iaccarino
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Andrew March
- Center for Research and Innovation, American College of Radiology, Reston, Virginia, USA
| | - Nidhi S Mundada
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Jhony Mejia Perez
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Karen Smith
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Claire Yballa
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Barry A Siegel
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Charles Windon
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
- Global Brain Health Institute, University of California San Francisco, San Francisco, California, USA
| | - Rachel A Whitmer
- Division of Research, Kaiser Permanente, Division of Research, Pleasanton, California, USA
- Department of Public Health Sciences, University of California Davis, Davis, California, USA
| | - Casaletto Kaitlin
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
- Global Brain Health Institute, University of California San Francisco, San Francisco, California, USA
| | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
- Department of Radiology & Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
2
|
Dasadhikari S, Ghosh S, Pal S, Knowles TPJ, Garai K. A single fibril study reveals that ApoE inhibits the elongation of Aβ42 fibrils in an isoform-dependent manner. Commun Chem 2025; 8:133. [PMID: 40307479 PMCID: PMC12044155 DOI: 10.1038/s42004-025-01524-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 04/15/2025] [Indexed: 05/02/2025] Open
Abstract
ApoE-ε4 is the strongest genetic risk factor for late-onset Alzheimer's disease (AD), linked to increased amyloid-β (Aβ) deposition in the brain. In AD mouse models, microglial expression of apoE3 reduces amyloid plaque burden through enhanced phagocytosis, whereas apoE4 is associated with impaired Aβ clearance. However, the isoform-specific interactions of apoE with Aβ aggregates and the molecular mechanisms by which these isoforms influence Aβ aggregation and clearance remain poorly understood, which is critical for developing potential therapeutic interventions. Here, we employed TIRFM, superresolution microscopy, and single-molecule photobleaching techniques to investigate the isoform-specific effects of apoE on the rate constants of Aβ42 aggregation at the single-fibril level, as well as to quantify the binding affinity and specificity of apoE isoforms to individual Aβ fibril ends. Our results show that apoE4 is ca. 4-5 times less effective than apoE3 and apoE2 in inhibiting fibril elongation, while secondary nucleation is largely unaffected by any of the isoforms. Furthermore, apoE3 exhibits stronger and more specific binding to fibril ends compared to apoE4. These findings suggest that apoE4's reduced affinity for growing fibril ends may impair microglial clearance and increase amyloid deposition through a higher elongation rate in the brain of ApoE-ε4 carriers.
Collapse
Affiliation(s)
| | - Shamasree Ghosh
- TIFR Centre for Interdisciplinary Sciences, Hyderabad, 500046, India
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, SE-90187, Sweden
| | - Sudip Pal
- TIFR Centre for Interdisciplinary Sciences, Hyderabad, 500046, India
| | - Tuomas P J Knowles
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
- Cavendish Laboratory, University of Cambridge, Cambridge, CB3 0HE, UK
| | - Kanchan Garai
- TIFR Centre for Interdisciplinary Sciences, Hyderabad, 500046, India.
| |
Collapse
|
3
|
Patel M, Pottier C, Fan KH, Cetin A, Johnson M, Ali M, Liu M, Gorijala P, Budde J, Shi R, Cohen AD, Becker JT, Snitz BE, Aizenstein H, Lopez OL, Morris JC, Kamboh MI, Cruchaga C. Whole-genome sequencing reveals the impact of lipid pathway and APOE genotype on brain amyloidosis. Hum Mol Genet 2025; 34:739-748. [PMID: 39927718 PMCID: PMC11973900 DOI: 10.1093/hmg/ddaf017] [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: 08/08/2024] [Revised: 10/11/2024] [Accepted: 01/29/2025] [Indexed: 02/11/2025] Open
Abstract
Amyloid-PET imaging tracks the accumulation of amyloid beta (Aβ) deposits in the brain. Amyloid plaques accumulation may begin 10 to 20 years before the individual experiences clinical symptoms associated with Alzheimer's diseases (ad). Recent large-scale genome-wide association studies reported common risk factors associated with brain amyloidosis, suggesting that this endophenotype is driven by genetic variants. However, these loci pinpoint to large genomic regions and the functional variants remain to be identified. To identify new risk factors associated with brain amyloid deposition, we performed whole-genome sequencing on a large cohort of European descent individuals with amyloid PET imaging data (n = 1,888). Gene-based analysis for coding variants was performed using SKAT-O for amyloid PET as a quantitative endophenotype that identified genome-wide significant association for APOE (P = 2.45 × 10-10), and 26 new candidate genes with suggestive significance association (P < 5. 0 × 10-03) including SCN7A (P = 7.31 × 10-05), SH3GL1 (P = 7.56 × 10-04), and MFSD12 (P = 8.51 × 10-04). Enrichment analysis highlighted the lipid binding pathways as associated with Aβ deposition in brain driven by PITPNM3 (P = 4.27 × 10-03), APOE (P = 2.45 × 10-10), AP2A2 (P = 1.06 × 10-03), and SH3GL1 (P = 7.56 × 10-04). Overall, our data strongly support a connection between lipid metabolism and the deposition of Aβ in the brain. Our study illuminates promising avenues for therapeutic interventions targeting lipid metabolism to address brain amyloidosis.
Collapse
Affiliation(s)
- Maulikkumar Patel
- Department of Psychiatry, Neurogenomics and Informatics, Washington University School of Medicine, 4444 Forest Park Ave, St. Louis, MO 63108, United States
| | - Cyril Pottier
- Department of Psychiatry, Neurogenomics and Informatics, Department of Neurology, Washington University School of Medicine, 4444 Forest Park Ave, St. Louis, MO 63108, United States
| | - Kang-Hsien Fan
- Department of Human Genetics, University of Pittsburgh, 130 De Soto St, Pittsburgh, PA 15261, United States
| | - Arda Cetin
- Department of Psychiatry, Neurogenomics and Informatics, Washington University School of Medicine, 4444 Forest Park Ave, St. Louis, MO 63108, United States
| | - Matthew Johnson
- Department of Psychiatry, Neurogenomics and Informatics, Washington University School of Medicine, 4444 Forest Park Ave, St. Louis, MO 63108, United States
| | - Muhammad Ali
- Department of Psychiatry, Neurogenomics and Informatics, Washington University School of Medicine, 4444 Forest Park Ave, St. Louis, MO 63108, United States
| | - Menghan Liu
- Department of Psychiatry, Neurogenomics and Informatics, Washington University School of Medicine, 4444 Forest Park Ave, St. Louis, MO 63108, United States
| | - Priyanka Gorijala
- Department of Psychiatry, Neurogenomics and Informatics, Washington University School of Medicine, 4444 Forest Park Ave, St. Louis, MO 63108, United States
| | - John Budde
- Department of Psychiatry, Neurogenomics and Informatics, Washington University School of Medicine, 4444 Forest Park Ave, St. Louis, MO 63108, United States
| | - Ruyu Shi
- Department of Human Genetics, University of Pittsburgh, 130 De Soto St, Pittsburgh, PA 15261, United States
| | - Ann D Cohen
- Department of Psychiatry, University of Pittsburgh, 3811 O'Hara Street, Pittsburgh, PA 15213, United States
| | - James T Becker
- Department of Neurology, University of Pittsburgh, 3471 Fifth Avenue, Pittsburgh, PA 15213, United States
| | - Beth E Snitz
- Department of Neurology, University of Pittsburgh, 3471 Fifth Avenue, Pittsburgh, PA 15213, United States
| | - Howard Aizenstein
- Department of Human Genetics, University of Pittsburgh, 130 De Soto St, Pittsburgh, PA 15261, United States
| | - Oscar L Lopez
- Department of Neurology, University of Pittsburgh, 3471 Fifth Avenue, Pittsburgh, PA 15213, United States
| | - John C Morris
- Department of Neurology, Hope Center for Neurologic Diseases, Section on Aging & Dementia, Institute of Clinical and Translational Sciences, Knight Alzheimer Disease Research Center Washington University School of Medicine, 4901 Forest Park Ave 4th floor, St. Louis, MO 63108, United States
| | - M Ilyas Kamboh
- Department of Human Genetics, Department of Psychiatry University of Pittsburgh, 130 De Soto St, Pittsburgh, PA 15261, United States
| | - Carlos Cruchaga
- Department of Psychiatry, Neurogenomics and Informatics, Department of Neurology, Hope Center for Neurologic Diseases, Knight Alzheimer Disease Research Center, Washington University School of Medicine, 4444 Forest Park Ave, St. Louis, MO 63108, United States
| |
Collapse
|
4
|
Jia B, Xu Y, Zhu X. Cognitive resilience in Alzheimer's disease: Mechanism and potential clinical intervention. Ageing Res Rev 2025; 106:102711. [PMID: 40021093 DOI: 10.1016/j.arr.2025.102711] [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: 01/02/2025] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
Alzheimer's disease (AD) is a globally recognized neurodegenerative disorder that severely impairs cognitive function and imposes substantial psychological and financial burdens on patients and their families. The hallmark pathological features of AD include progressive neurodegeneration, extracellular beta-amyloid (Aβ) plaque accumulation, and intracellular hyperphosphorylated tau protein tangles. However, recent studies have identified a subset of patients exhibiting cognitive resilience, characterized by a slower cognitive decline or the preservation of high cognitive function despite the presence of AD pathology. Cognitive resilience is influenced by a complex interplay of genetic, environmental, and lifestyle factors. In addition, cognitive resilience contributes to the new perspectives on the diagnosis and personalized treatment of AD. This review aims to provide a comprehensive analysis of current studies on cognitive resilience in AD and to explore future research directions of AD diagnosis and treatment.
Collapse
Affiliation(s)
- Bin Jia
- Department of Neurology, Nanjing Drum Tower Hospital, School of Medicine, Jiangsu University, Nanjing, Jiangsu, China; Department of Neurology, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, School of Medicine, Jiangsu University, Nanjing, Jiangsu, China; Jiangsu Key Laboratory for Molecular Medicine and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China; Nanjing Neurology Clinical Medical Center, and Nanjing Drum Tower Hospital Brain Disease and Brain Science Center, Nanjing, China
| | - Xiaolei Zhu
- Department of Neurology, Nanjing Drum Tower Hospital, School of Medicine, Jiangsu University, Nanjing, Jiangsu, China; Jiangsu Key Laboratory for Molecular Medicine and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China; Nanjing Neurology Clinical Medical Center, and Nanjing Drum Tower Hospital Brain Disease and Brain Science Center, Nanjing, China.
| |
Collapse
|
5
|
Tanure YCB, Mafra ACM, Guimarães BLM, Magalhães RC, Fagundez C, Nascimento IJBD, Brito JCM. Potential benefits of kefir and its compounds on Alzheimer's disease: A systematic review. BRAIN BEHAVIOR AND IMMUNITY INTEGRATIVE 2025; 10:100115. [PMID: 40376196 PMCID: PMC12075057 DOI: 10.1016/j.bbii.2025.100115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/24/2025] [Accepted: 03/12/2025] [Indexed: 05/18/2025]
Abstract
Alzheimer's disease, characterized by the progressive loss of cognitive functions of the brain, is still an incurable pathology. Current treatments primarily aim to alleviate symptoms, acting mainly on behavioral changes, having a modest impact in the disease course. Recently, potential role of probiotics in managing Alzheimer's has been explored. Kefir, a fermented food teeming with live microorganisms, is thought to influence the gut microbiota, potentially reducing inflammation and the accumulation of toxic proteins in the brain. Additionally, kefir contains bioactive compounds, such as B vitamins, choline, and folic acid, which are essential for neuronal health and cognitive function. Thus, kefir could emerge as a promising complementary treatment for Alzheimer's disease. This systematic review, conducted in January 2024, examined the effects of kefir in both in vivo animal models and human patients with neurodegenerative conditions. The review was based on studies retrieved from BVS, Embase, PubMed/MEDLINE, Scopus, and Web of Science databases. Seven studies were included, involving invertebrates, murine models, and human participants. In animal models, the primary outcomes were antioxidant effects, reduced beta-amyloid deposition, and attenuation of vascular damage and neurodegeneration. In human studies, kefir supplementation resulted in decreased levels of inflammatory cytokines, reactive oxygen species (ROS), and oxidative proteins, and was associated with improvements in memory. Given its potential benefits, kefir could serve as a valuable adjunct to conventional treatments for Alzheimer's disease, warranting further investigation in clinical settings.
Collapse
|
6
|
Choi M, Zimmerman SC, Buto PT, Wang J, Brenowitz WD, Hoffmann TJ, Hazzouri AZA, Kezios K, Glymour MM. Association of genetic risk score for Alzheimer's disease with late-life body mass index in all of us: Evaluating reverse causation. Alzheimers Dement 2025; 21:e14598. [PMID: 40189781 PMCID: PMC11972977 DOI: 10.1002/alz.14598] [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: 09/20/2024] [Revised: 01/03/2025] [Accepted: 01/14/2025] [Indexed: 04/10/2025]
Abstract
INTRODUCTION Decreases in body mass index (BMI) may be early consequences of Alzheimer's disease (AD) pathophysiological changes. Previous research in the UK Biobank estimated that AD-related genes began affecting BMI around age 47. We assessed whether this result could be replicated using longitudinal data in an independent cohort. METHODS Using All of Us (AOU) (N = 197,619, aged 30+) data, we estimated linear mixed models for associations of Z-scored AD-Genetic Risk Score (AD-GRS) with BMI, stratified by decade of age. We calculated the earliest age at which AD-GRS was associated with differences in BMI using cross-validated models adjusted for demographics. RESULTS Higher AD-GRS was statistically associated with lower BMI in participants aged 60-70 (b = -0.060 [-0.113, -0.007]). Best fitting models suggested the inverse association of AD-GRS and BMI emerged beginning at ages 47-54. DISCUSSION AD genes accelerate age-related weight loss starting in middle age. HIGHLIGHTS Understanding when physiological changes from amyloid pathology begin is key for AD prevention. Our findings indicate that AD-associated genes accelerate midlife weight loss, starting between 47 and 54 years. AD prevention research should consider that disease pathology likely begins by middle age.
Collapse
Affiliation(s)
- Minhyuk Choi
- Department of Epidemiology and BiostatisticsUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Scott C. Zimmerman
- Department of Epidemiology and BiostatisticsUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Peter T. Buto
- Department of Epidemiology and BiostatisticsUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of EpidemiologyBoston University School of Public HealthBostonMassachusettsUSA
| | - Jingxuan Wang
- Department of Epidemiology and BiostatisticsUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Willa D. Brenowitz
- Department of Epidemiology and BiostatisticsUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Kaiser Permanente Center for Health ResearchPortlandOregonUSA
| | - Thomas J. Hoffmann
- Department of Epidemiology and BiostatisticsUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Institute for Human GeneticsUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | | | - Katrina Kezios
- Department of EpidemiologyColumbia UniversityNew YorkNew YorkUSA
| | - M. Maria Glymour
- Department of EpidemiologyBoston University School of Public HealthBostonMassachusettsUSA
| |
Collapse
|
7
|
Xue RJ, Gao PY, Chen YM, Liu Y, Han BL, Huang YM, Mi YC, Cui RP, Lin YJ, Wang ZT, Tan CC, Ou YN, Tan L. Associations between plasma complement C1q and cerebrospinal fluid biomarkers of Alzheimer's disease pathology in cognitively intact adults: The CABLE study. J Alzheimers Dis 2025; 104:1136-1146. [PMID: 40138515 DOI: 10.1177/13872877251322808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
BackgroundC1q is a promoter of the classical pathway of complement and its massive expression may be associated with the development of Alzheimer's disease (AD). However, the relationships between C1q and the major pathological challenges, including amyloid-β (Aβ) and tau deposition, remain undetermined in the preclinical AD phase.ObjectiveThis study aims to investigate the connections between plasma C1q and CSF AD biomarkers.MethodsThe cognitively intact participants (N = 1264) from the Chinese Alzheimer's Biomarker and LifestylE (CABLE) study were categorized into four groups, including Stage 0 [normal Amyloid-β1-42 (Aβ1-42), Phosphorylated-tau (P-tau) and Total-tau (T-tau)], Stage 1 (abnormal Aβ1-42, but normal P-tau or T-tau), Stage 2 (abnormal Aβ1-42 and abnormal P-tau or T-tau), and suspected non-Alzheimer disease pathology (SNAP) (abnormal P-tau or T-tau, but normal amyloid levels). The changes in plasma C1q levels among these groups and the correlation between C1q levels and cerebrospinal fluid (CSF) AD biomarkers were performed.ResultsThe results demonstrated plasma C1q levels are lower in Stage 0 (p = 0.010) and SNAP (p < 0.001) compared with Stage 1. A significant association between C1q levels and CSF AD pathology, including Aβ1-42 (β = -0.143, p < 0.001), Aβ1-42/Aβ1-40 (β = -0.173, p < 0.001), P-tau/Aβ1-42 (β = 0.156, p < 0.001), and T-tau/Aβ1-42 (β = 0.130, p < 0.001) has been identified.ConclusionsThe current research elucidates a positive correlation between elevated plasma C1q levels and CSF Aβ pathology, with C1q amplifying concomitantly with the pathological and clinical progression of AD.
Collapse
Affiliation(s)
- Rong-Ji Xue
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Pei-Yang Gao
- Department of Neurology & Innovation Center for Neurological Disorders, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, China
| | - Yan-Ming Chen
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ying Liu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Bao-Lin Han
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yi-Ming Huang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yin-Chu Mi
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China
| | - Rui-Ping Cui
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yu-Jing Lin
- Department of Neurology, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Zuo-Teng Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Chen-Chen Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Nan Ou
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| |
Collapse
|
8
|
Andrey T, Alexander S, Inna B, Daria Z, Viktoria A, Anastasiia SG, Kumar A, Ivan F, Arina E, Oxana SG. Age as a limiting factor for effectiveness of photostimulation of brain drainage and cognitive functions. FRONTIERS OF OPTOELECTRONICS 2025; 18:6. [PMID: 40163163 PMCID: PMC11958890 DOI: 10.1007/s12200-025-00149-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 01/21/2025] [Indexed: 04/02/2025]
Abstract
The progressive number of old adults with cognitive impairment worldwide and the lack of effective pharmacologic therapies require the development of non-pharmacologic strategies. The photobiomodulation (PBM) is a promising method in prevention of early or mild age-related cognitive impairments. However, it remains unclear the efficacy of PBM for old patients with significant age-related cognitive dysfunction. In our study on male mice, we show a gradual increase in the brain amyloid beta (Aβ) levels and a decrease in brain drainage with age, which, however, is associated with a decline in cognitive function only in old (24 months of age) mice but not in middle-aged (12 months of age) and young (3 month of age) animals. These age-related features are accompanied by the development of hyperplasia of the meningeal lymphatic vessels (MLVs) in old mice underlying the decrease in brain drainage. PBM improves cognitive training exercises and Aβ clearance only in young and middle-aged mice, while old animals are not sensitive to PBM. These results clearly demonstrate that the PBM effects on cognitive function are correlated with age-mediated changes in the MLV network and may be effective if the MLV function is preserved. These findings expand fundamental knowledge about age differences in the effectiveness of PBM for improvement of cognitive functions and Aβ clearance as well as about the lymphatic mechanisms responsible for age decline in sensitivity to the therapeutic PBM effects.
Collapse
Affiliation(s)
- Terskov Andrey
- Department of Biology, Saratov State University, Saratov, 410012, Russia
| | - Shirokov Alexander
- Department of Biology, Saratov State University, Saratov, 410012, Russia
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Russian Academy of Sciences, Saratov, 410049, Russia
| | - Blokhina Inna
- Department of Biology, Saratov State University, Saratov, 410012, Russia
| | | | - Adushkina Viktoria
- Department of Biology, Saratov State University, Saratov, 410012, Russia
| | | | - Atul Kumar
- The Indian Institute of Technology (BHU) Varanasi, Uttar Pradesh, Varanasi, 221005, India
| | - Fedosov Ivan
- Institute of Physics, Saratov State University, Saratov, 410012, Russia
| | - Evsukova Arina
- Department of Biology, Saratov State University, Saratov, 410012, Russia
| | | |
Collapse
|
9
|
Govindarajan ST, Mamourian E, Erus G, Abdulkadir A, Melhem R, Doshi J, Pomponio R, Tosun D, Bilgel M, An Y, Sotiras A, Marcus DS, LaMontagne P, Benzinger TLS, Espeland MA, Masters CL, Maruff P, Launer LJ, Fripp J, Johnson SC, Morris JC, Albert MS, Bryan RN, Resnick SM, Habes M, Shou H, Wolk DA, Nasrallah IM, Davatzikos C. Machine learning reveals distinct neuroanatomical signatures of cardiovascular and metabolic diseases in cognitively unimpaired individuals. Nat Commun 2025; 16:2724. [PMID: 40108173 PMCID: PMC11923046 DOI: 10.1038/s41467-025-57867-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 03/03/2025] [Indexed: 03/22/2025] Open
Abstract
Comorbid cardiovascular and metabolic risk factors (CVM) differentially impact brain structure and increase dementia risk, but their specific magnetic resonance imaging signatures (MRI) remain poorly characterized. To address this, we developed and validated machine learning models to quantify the distinct spatial patterns of atrophy and white matter hyperintensities related to hypertension, hyperlipidemia, smoking, obesity, and type-2 diabetes mellitus at the patient level. Using harmonized MRI data from 37,096 participants (45-85 years) in a large multinational dataset of 10 cohort studies, we generated five in silico severity markers that: i) outperformed conventional structural MRI markers with a ten-fold increase in effect sizes, ii) captured subtle patterns at sub-clinical CVM stages, iii) were most sensitive in mid-life (45-64 years), iv) were associated with brain beta-amyloid status, and v) showed stronger associations with cognitive performance than diagnostic CVM status. Integrating personalized measurements of CVM-specific brain signatures into phenotypic frameworks could guide early risk detection and stratification in clinical studies.
Collapse
Grants
- RF1 AG054409 NIA NIH HHS
- U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- The iSTAGING study is a multi-institutional effort funded by the National Institute on Aging (NIA) by RF1 AG054409 (C. Davatzikos). Data used in preparation of this article were obtained from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) database (adni.loni.usc.edu). As such, the investigators within the ADNI contributed to the design and implementation of ADNI and/or provided data but did not participate in analysis or writing of this report. A complete listing of ADNI investigators can be found at: http://adni.loni.usc.edu/wp-content/uploads/how_to_apply/ADNI_Acknowledgement_List.pdf. ADNI is funded by the NIA, the National Institute of Biomedical Imaging and Bioengineering and through generous contributions from the following: AbbVie, Alzheimer’s Association; Alzheimer’s Drug Discovery Foundation; Araclon Biotech; BioClinica; Biogen; Bristol-Myers Squibb; CereSpir; Cogstate; Eisai; Elan Pharmaceuticals; Eli Lilly and Company; EuroImmun; F. Hoffmann-La Roche and its affiliated company Genentech; Fujirebio; GE Healthcare; IXICO; Janssen Alzheimer Immunotherapy Research & Development; Johnson & Johnson Pharmaceutical Research & Development; Lumosity; Lundbeck; Merck & Co; Meso Scale Diagnostics; NeuroRx Research; Neurotrack Technologies; Novartis Pharmaceuticals Corporation; Pfizer; Piramal Imaging; Servier; Takeda Pharmaceutical Company; and Transition Therapeutics. The Canadian Institutes of Health Research is providing funds to support ADNI clinical sites in Canada. Private sector contributions are facilitated by the Foundation for the National Institutes of Health (www.fnih.org). The grantee organization is the Northern California Institute for Research and Education, and the study is coordinated by the Alzheimer’s Therapeutic Research Institute at the University of Southern California. ADNI data are disseminated by the Laboratory for Neuro Imaging at the University of Southern California. Data used in the preparation of this article was obtained from the Australian Imaging Biomarkers and Lifestyle flagship study of ageing (AIBL) funded by the Commonwealth Scientific and Industrial Research Organisation (CSIRO) which was made available at the ADNI database (www.loni.usc.edu/ADNI). The AIBL researchers contributed data but did not participate in analysis or writing of this report. AIBL researchers are listed at www.aibl.csiro.au. The BIOCARD study is partly supported by NIH grant U19-AG033655 (M.S. Albert). The BLSA neuroimaging study is funded by the Intramural Research Program, NIA, National Institutes of Health (NIH), and by HHSN271201600059C (S. M. Resnick, M. Bilgel, Y. An). CARDIA study is conducted and supported by the NHLBI in collaboration with the University of Alabama at Birmingham (HHSN268201300025C and HHSN268201300026C), Northwestern University (HHSN268201300027C), University of Minnesota (HHSN268201300028C), Kaiser Foundation Research Institute (HHSN268201300029C), and Johns Hopkins University School of Medicine (HHSN268200900041C). CARDIA is also partially supported by the Intramural Research Program of the National Institute on Aging (NIA) and an intra-agency agreement between NIA and NHLBI (AG0005) (L.J. Launer). Data used in the preparation of this article was obtained from the OASIS study funded in part by grants P50 AG05681, P01 AG03991, P01 AG026276, R01 AG021910, P20 MH071616, U24 RR021382 for OASIS-1, P50 AG05681, P01 AG03991, P01 AG026276, R01 AG021910, P20 MH071616, U24 RR021382 for OASIS-2, and NIH P30 AG066444, P50 AG00561, P30 NS09857781, P01 AG026276, P01 AG003991, R01 AG043434, UL1 TR000448, R01 EB009352 for OASIS-3 (T. Benzinger, D. Marcus, J. Morris, P. LaMontagne). Data used in the preparation of this article was obtained at Penn Alzheimer’s Disease Research Center funded in part by grant P30 AG072979 (D.A. Wolk). Data used in the preparation of this article was obtained from the UK Biobank Resource under application number 35148. The Women’s Health Initiative was funded by the National Heart, Lung and Blood Institute of the NIH, US Department of Health and Human Services. Contracts HHSN268200464221C and N01-WH-4-4221 provided additional support. The WHIMS (M.A. Espeland) was funded in part by Wyeth Pharmaceuticals. The WRAP study was supported by grants: NIH R01AG027161 and R01AG054047 (S.C. Johnson). The authors would like to acknowledge the clinical and neuropathology diagnostic support provided by the Wisconsin ADRC’s Clinical, Neuropathology and Biomarkers Cores, and biostatistical support provided by the Data Management and Biostatistics Core. S.T. Govindarajan was partly supported by the Alzheimer’s Association Research Fellowship AARFD-23-1151286. A. Abdulkadir was funded through grants 191026 and 206795 awarded by the Swiss National Science Foundation. M. Habes was supported by grant 1R01AG080821 from the National Institutes of Health.
Collapse
Affiliation(s)
| | - Elizabeth Mamourian
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Guray Erus
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Ahmed Abdulkadir
- Centre for Artificial Intelligence, ZHAW School of Engineering, Winterthur, Switzerland
| | - Randa Melhem
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Jimit Doshi
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Raymond Pomponio
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Murat Bilgel
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Aristeidis Sotiras
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel S Marcus
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Pamela LaMontagne
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Mark A Espeland
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Colin L Masters
- Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Paul Maruff
- Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Lenore J Launer
- Neuroepidemiology Section, Intramural Research Program, National Institute on Aging, Bethesda, MD, USA
| | - Jurgen Fripp
- CSIRO Health and Biosecurity, Australian e-Health Research Centre CSIRO, Brisbane, Queensland, Australia
| | - Sterling C Johnson
- Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Marilyn S Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - R Nick Bryan
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Mohamad Habes
- Biggs Alzheimer's Institute, University of Texas San Antonio Health Science Center, San Antonio, TX, USA
| | - Haochang Shou
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - David A Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Ilya M Nasrallah
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
10
|
DeCarli C, Rajan KB, Jin LW, Hinman J, Johnson DK, Harvey D, Fornage M, on behalf of the Diverse Vascular Contributions to Cognitive Impairment and Dementia (Diverse VCID) Study Investigators. WMH Contributions to Cognitive Impairment: Rationale and Design of the Diverse VCID Study. Stroke 2025; 56:758-776. [PMID: 39545328 PMCID: PMC11850211 DOI: 10.1161/strokeaha.124.045903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
As awareness of dementia increases, more individuals with minor cognitive complaints are requesting clinical assessment. Neuroimaging studies frequently identify incidental white matter hyperintensities, raising patient concerns about their brain health and future risk for dementia. Moreover, current US demographics indicate that ≈50% of these individuals will be from diverse backgrounds by 2060. Racial and ethnic minority populations bear a disproportionate burden of vascular risk factors magnifying dementia risk. Despite established associations between white matter hyperintensities and cognitive impairment, including dementia, no study has comprehensively and prospectively examined the impact of individual and combined magnetic resonance imaging measures of white matter injury, their risk factors, and comorbidities on cognitive performance among a diverse, nondemented, stroke-free population with cognitive complaints over an extended period of observation. The Diverse VCID (Diverse Vascular Cognitive Impairment and Dementia) study is designed to fill this knowledge gap through 3 assessments of clinical, behavioral, and risk factors; neurocognitive and magnetic resonance imaging measures; fluid biomarkers of Alzheimer disease, vascular inflammation, angiogenesis, and endothelial dysfunction; and measures of genetic risk collected prospectively over a minimum of 3 years in a cohort of 2250 individuals evenly distributed among Americans of Black/African, Latino/Hispanic, and non-Hispanic White backgrounds. The goal of this study is to investigate the basic mechanisms of small vessel cerebrovascular injury, emphasizing clinically relevant assessment tools and developing a risk score that will accurately identify at-risk individuals for possible treatment or clinical therapeutic trials, particularly individuals of diverse backgrounds where vascular risk factors and disease are more prevalent.
Collapse
Affiliation(s)
- Charles DeCarli
- Department of Neurology, University of California at Davis, Sacramento, CA, USA
| | - Kumar B. Rajan
- Rush Institute for Healthy Aging, Rush University Medical Center, Chicago IL
| | - Lee-Way Jin
- Department of Pathology and Laboratory Medicine University of California Davis California USA
| | - Jason Hinman
- Department of Neurology, University of California, Los Angeles, Los Angeles, CA, United States
| | - David K. Johnson
- Department of Neurology, University of California at Davis, Sacramento, CA, USA
| | - Danielle Harvey
- Department of Public Health Sciences University of California Davis California USA
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | | |
Collapse
|
11
|
Joutsa J, Rinne JO, Niemi KJ, Karrasch M, Parkkola RK, Saunavaara J, Helin SP, Hermann BP, Sillanpää M. Progression of Amyloid Accumulation in Late Adulthood Among People With Childhood-Onset Epilepsy. Neurology 2025; 104:e210303. [PMID: 39823561 DOI: 10.1212/wnl.0000000000210303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 11/27/2024] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Previous research has demonstrated increased brain amyloid plaque load in individuals with childhood-onset epilepsy in late middle age. However, the trajectory of this process is not yet known. The aim of this study was to determine whether individuals with a history of childhood-onset epilepsy show progressive brain aging in amyloid accumulation in late adulthood (Turku Adult Childhood-Onset Epilepsy study, TACOE). METHODS Adults from a prospective population-based cohort of individuals with childhood-onset epilepsy, originally recruited 1961-1964, together with matched controls, were scanned with [11C]PIB PET twice: after at least 50 years (TACOE-50) and again after at least 55 years (TACOE-55) from the diagnosis. RESULTS At TACOE-55, 31.4% (11/36, mean age 63.3 years, 52.8% female) of individuals from the epilepsy group and 11.4% (4/35, 63.1 year, 54.3%) of controls had a visually abnormal [11C]PIB scan (p = 0.039). At TACOE-55, cortical brain [11C]PIB uptakes were higher and increased more from TACOE-50 in the epilepsy compared with the control group (p < 0.05). In voxelwise whole-brain analyses, the epilepsy group showed significantly higher and more widespread brain amyloid accumulation (pFWE < 0.05). DISCUSSION The results demonstrate that childhood-onset epilepsy is associated with an earlier age at onset of amyloidosis and greater progressive amyloid accumulation in late adulthood.
Collapse
Affiliation(s)
- Juho Joutsa
- Turku Brain and Mind Center, Clinical Neurosciences, University of Turku, Finland
- Neurocenter, Turku University Hospital, Finland
- Turku PET Centre, University of Turku, Finland
| | - Juha O Rinne
- Neurocenter, Turku University Hospital, Finland
- Turku PET Centre, University of Turku, Finland
| | - Kalle J Niemi
- Turku Brain and Mind Center, Clinical Neurosciences, University of Turku, Finland
- Neurocenter, Turku University Hospital, Finland
| | - Mira Karrasch
- Department of Psychology, Åbo Akademi, Turku, Finland
| | - Riitta K Parkkola
- Department of Radiology, University of Turku and Turku University Hospital, Finland
| | - Jani Saunavaara
- Department of Medical Physics, Turku University Hospital and University of Turku, Finland
| | | | - Bruce P Hermann
- Department of Neurology, University of Wisconsin School of Medicine and Public Health, Madison, WI; and
| | - Matti Sillanpää
- Departments of Child Neurology and General Practice, University of Turku and Turku University Hospital, Finland
| |
Collapse
|
12
|
Cacciaglia R, Falcón C, Benavides GS, Brugulat‐Serrat A, Alomà MM, Calvet MS, Molinuevo JL, Fauria K, Minguillón C, Kollmorgen G, Quijano‐Rubio C, Blennow K, Zetterberg H, Lorenzini L, Wink AM, Ingala S, Barkhof F, Ritchie CW, Gispert JD, for the ALFA study. Soluble Aβ pathology predicts neurodegeneration and cognitive decline independently on p-tau in the earliest Alzheimer's continuum: Evidence across two independent cohorts. Alzheimers Dement 2025; 21:e14415. [PMID: 39898436 PMCID: PMC11848178 DOI: 10.1002/alz.14415] [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: 05/01/2024] [Revised: 10/07/2024] [Accepted: 10/27/2024] [Indexed: 02/04/2025]
Abstract
INTRODUCTION Identifying the link between early Alzheimer's disease (AD) pathological changes and neurodegeneration in asymptomatic individuals may lead to the discovery of preventive strategies. We assessed longitudinal brain atrophy and cognitive decline as a function of cerebrospinal fluid (CSF) AD biomarkers in two independent cohorts of cognitively unimpaired (CU) individuals. METHODS We used longitudinal voxel-based morphometry (VBM) in combination with hippocampal subfield segmentation. Changes in neuroimaging and cognitive variables were inspected using general linear models (GLMs) adjusting by age, sex, apolipoprotein E (APOE) status, follow-up time, and years of education. RESULTS In both cohorts, baseline CSF amyloid beta (Aβ) biomarkers significantly predicted medial temporal lobe (MTL) atrophy rates and episodic memory (EM) decline independently of CSF phosphorylated tau (p-tau). DISCUSSION Our data suggest that soluble Aβ dyshomeostasis triggers MTL longitudinal atrophy and EM decline independently of CSF p-tau. Our data underscore the need for secondary preventive strategies at the earliest stages of the AD pathological cascade. HIGHLIGHTS We assessed brain atrophy and cognitive decline in asymptomatic individuals. Aβ biomarkers predicted MTL atrophy independently of p-tau. Our results underscore the importance of undertaking Alzheimer's preclinical trials.
Collapse
Grants
- #ALFGBG-715986 the Swedish state under the agreement between the Swedish government and the County Councils, the Avtal om Läkarutbildning och Forskning (ALF)-agreement
- #RDAPB-201809-2016615 the Alzheimer Drug Discovery Foundation (ADDF), USA
- #AF-968270 the Swedish Alzheimer Foundation
- JPND2021-00694 the European Union Joint Programme - Neurodegenerative Disease Research
- Project "PI19/00155" European Union's Horizon 2020 Research and Innovation Programme (Grant agreement No. 948677)
- No. 101053962 the European Union's Horizon Europe Research and Innovation Programme under Grant Agreement
- #FO2017-0243 Hjärnfonden, Sweden
- ZEN-21-848495 the Alzheimer's Association 2021 Zenith Award
- #2018-02532 HZ is a Wallenberg Scholar supported by grants from the Swedish Research Council
- MSC receives funding from the European Research Council (ERC)
- #ALZ2022-0006 Hjärnfonden, Sweden
- #AF-939721 the Swedish Alzheimer Foundation
- the European Union Next Generation EU/Plan de Recuperación
- #ADSF-21-831377-C the AD Strategic Fund and the Alzheimer's Association
- MCIN/AEI/10.13039/501100011033/FEDER RC receives funding from "Ministerio de Ciencia, Innovación y Universidades - Agencia Estatal de Investigación"
- PID2021-125433OA-100 RC receives funding from "Ministerio de Ciencia, Innovación y Universidades - Agencia Estatal de Investigación"
- Transformación y Resiliencia (PRTR)
- LCF/BQ/PR21/11840004 the European Union's Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie grant agreement No 847648
- the Bluefield Project, the Olav Thon Foundation
- SG-23-1038904 QC the Alzheimer's Association 2022-2025
- R01 AG068398 NIA NIH HHS
- MCIN/AEI/10.13039/501100011033 RC receives funding from "Ministerio de Ciencia, Innovación y Universidades - Agencia Estatal de Investigación"
- #ALFGBG-965240 the Swedish state under the agreement between the Swedish government and the County Councils, the Avtal om Läkarutbildning och Forskning (ALF)-agreement
- #FO2022-0270 the Erling-Persson Family Foundation, Stiftelsen för Gamla Tjänarinnor, Hjärnfonden, Sweden
- the European Union's Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie grant agreement No. 860197 (MIRIADE)
- JPND2019-466-236 the European Union Joint Program for Neurodegenerative Disorders
- #1R01AG068398-01 the National Institute of Health (NIH), USA
- UKDRI-1003 the UK Dementia Research Institute at University College London (UCL)
- #ALFGBG-71320 Swedish State Support for Clinical Research
- #201809-2016862 the Alzheimer Drug Discovery Foundation (ADDF), USA
- #ADSF-21-831376-C the AD Strategic Fund and the Alzheimer's Association
- #AF-930351 the Swedish Alzheimer Foundation
- #2017-00915 KB is supported by the Swedish Research Council
- ID 100010434 Instituto de Salud Carlos III (ISCIII) and co-funded by the European Union, and from a fellowship from "la Caixa" Foundation
- #ADSF-21-831381-C the AD Strategic Fund and the Alzheimer's Association
- RYC2021-031128-I RC receives funding from "Ministerio de Ciencia, Innovación y Universidades - Agencia Estatal de Investigación"
Collapse
Affiliation(s)
- Raffaele Cacciaglia
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research InstituteBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
| | - Carles Falcón
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research InstituteBarcelonaSpain
- Centro de Investigación Biomédica en Red de BioingenieríaBiomateriales y Nanomedicina (CIBERBBN)MadridSpain
| | - Gonzalo Sánchez Benavides
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research InstituteBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
| | - Anna Brugulat‐Serrat
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research InstituteBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
- Global Brain Health InstituteSan FranciscoCaliforniaUSA
| | - Marta Milà Alomà
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall FoundationBarcelonaSpain
- Northern California Institute for Research and EducationSan FranciscoCaliforniaUSA
| | - Marc Suárez Calvet
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research InstituteBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
- Servei de NeurologiaHospital del MarBarcelonaSpain
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall FoundationBarcelonaSpain
- Present address:
Ottiliavej 9, 2500KøbenhavnDenmark
| | - Karine Fauria
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research InstituteBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
| | - Carolina Minguillón
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research InstituteBarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
| | | | | | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and PhysiologyThe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- UK Dementia Research Institute at UCLLondonUK
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Luigi Lorenzini
- Department of Radiology & Nuclear MedicineAmsterdam UMC, Vrije UniversiteitAmsterdamthe Netherlands
| | - Alle Meije Wink
- Department of Radiology & Nuclear MedicineAmsterdam UMC, Vrije UniversiteitAmsterdamthe Netherlands
| | - Silvia Ingala
- Department of Radiology & Nuclear MedicineAmsterdam UMC, Vrije UniversiteitAmsterdamthe Netherlands
| | - Frederik Barkhof
- Department of Radiology & Nuclear MedicineAmsterdam UMC, Vrije UniversiteitAmsterdamthe Netherlands
- Queen Square Institute of Neurology and Centre for Medical Image ComputingUniversity College LondonLondonUK
| | - Craig W. Ritchie
- Edinburgh Dementia Prevention, Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghScotlandUK
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Research InstituteBarcelonaSpain
- Centro de Investigación Biomédica en Red de BioingenieríaBiomateriales y Nanomedicina (CIBERBBN)MadridSpain
- Universitat Pompeu FabraBarcelonaSpain
| | | |
Collapse
|
13
|
Ou Z, Pan Y, Xie F, Guo Q, Shen D. Image-and-Label Conditioning Latent Diffusion Model: Synthesizing A$\beta$-PET From MRI for Detecting Amyloid Status. IEEE J Biomed Health Inform 2025; 29:1221-1231. [PMID: 40030191 DOI: 10.1109/jbhi.2024.3492020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2025]
Abstract
Deposition of $\beta$-amyloid (A$\beta$), which is generally observed by A$\beta$-PET, is an important biomarker to evaluate subjects with early-onset dementia. However, acquisition of A$\beta$-PET usually suffers from high expense and radiation hazards, making A$\beta$-PET not commonly used as MRI. As A$\beta$-PET scans are only used to determine whether A$\beta$ deposition is positive or not, it is highly valuable to capture the underlying relationship between A$\beta$ deposition and other neuroimages (i.e., MRI) and detect amyloid status based on other neuroimages to reduce necessity of acquiring A$\beta$-PET. To this end, we propose an image-and-label conditioning latent diffusion model (IL-CLDM) to synthesize A$\beta$-PET scans from MRI scans by enhancing critical shared information to finally achieve MRI-based A$\beta$ classification. Specifically, two conditioning modules are introduced to enable IL-CLDM to implicitly learn joint image synthesis and diagnosis: 1) an image conditioning module, to extract meaningful features from source MRI scans to provide structural information, and 2) a label conditioning module, to guide the alignment of generated scans to the diagnosed label. Experiments on a clinical dataset of 510 subjects demonstrate that our proposed IL-CLDM achieves image quality superior to five widely used models, and our synthesized A$\beta$-PET scans (by IL-CLDM) can significantly help classification of A$\beta$ as positive or negative.
Collapse
|
14
|
Patil S, Ayubcha C, Teichner E, Subtirelu R, Cho JH, Ghonim M, Ghonim M, Werner TJ, Høilund-Carlsen PF, Alavi A, Newberg AB. Clinical Applications of PET Imaging in Alzheimer's Disease. PET Clin 2025; 20:89-100. [PMID: 39547733 DOI: 10.1016/j.cpet.2024.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Alzheimer's disease (AD) involves a complex pathophysiology of neurodegeneration that leads to severe cognitive deficiencies. Understanding the molecular alterations that underlie this disease is fundamental to clinical management and therapeutic innovation. Functional imaging with positron emission tomography (PET) enables a visualization of these impaired pathways, such as cerebral hypometabolism, amyloid and tau accumulation, and neurotransmitter dysfunction. This review discusses the clinical applications of PET in AD and mild cognitive impairment, focusing on relevant original research studies for disease diagnosis, progression, and treatment response.
Collapse
Affiliation(s)
- Shiv Patil
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA; Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Cyrus Ayubcha
- Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Eric Teichner
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA; Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Robert Subtirelu
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA
| | | | - Mohanad Ghonim
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA; Department of Radiology, Ain Shams University, Cairo, Egypt
| | - Mohamed Ghonim
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA; Department of Radiology, Ain Shams University, Cairo, Egypt
| | - Thomas J Werner
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA
| | | | - Abass Alavi
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA
| | - Andrew B Newberg
- Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA 19107, USA; Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
15
|
Socher KLR, Nunes DM, Lopes DCP, Coutinho AMN, Faria DDP, Squarzoni P, Busatto Filho G, Buchpiguel CA, Nitrini R, Brucki SMD. Diagnosing preclinical and clinical Alzheimer's disease with visual atrophy scales in the clinical practice. ARQUIVOS DE NEURO-PSIQUIATRIA 2025; 83:1-7. [PMID: 40020758 DOI: 10.1055/s-0045-1802960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/03/2025]
Abstract
BACKGROUND Visual atrophy scales from the medial temporal region are auxiliary biomarkers of neurodegeneration in Alzheimer's disease (AD). Therefore, they may correlate with progression from cognitively unimpaired (CU) status to mild cognitive impairment (MCI) and AD, and they become a valuable tool for diagnostic accuracy. OBJECTIVE To compare the medial temporal lobe atrophy (MTA) and entorhinal cortex atrophy (ERICA) scores measured through magnetic resonance image (MRI) scans as a useful method for probable AD diagnosis regarding clinical diagnosis and amyloid positron-emission tomography (PET). METHODS Two neurologists blinded to the diagnoses classified 113 older adults (age > 65 years) through the MTA and ERICA scores. We investigated the correlations involving these scores and sociodemographic data, amyloid brain cortical burden measured through PET imaging with (11)C-labeled Pittsburgh Compound-B (11C-PIB PET), and clinical cognitive status, in individuals diagnosed as CU (CU; N = 30), presenting mild cognitive impairment (MCI, N = 52), and AD patients (N = 31). RESULTS The inter-rater reliability of the atrophy scales was excellent (0.8-1) according to the Cohen analysis. The CU group presented lower MTA scores (median value: 0) than ERICA (median value: 1) scores in both hemispheres. The 11C-PIB-PET was positive in 45% of the sample. In the MCI and AD groups, the ERICA score presented greater sensitivity, and the MTA score presented greater specificity. The accuracy of the clinical diagnosis was sufficient and no more than 70% for both scores in AD. CONCLUSION In the present study, we found moderate sensitivity for the ERICA score, which could be a better screening tool than the MTA score for the diagnosis of AD or MCI. However, none of the scores were useful imaging biomarkers in preclinical AD.
Collapse
Affiliation(s)
- Karen Luiza Ramos Socher
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| | - Douglas Mendes Nunes
- Universidade de São Paulo, Faculdade de Medicina, Instituto de Radiologia, Unidade de Neurorradiologia, São Paulo SP, Brazil
| | - Deborah Cristina P Lopes
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| | - Artur Martins Novaes Coutinho
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Radiologia e Oncologia, Laboratório de Medicina Nuclear (LIM/43), São Paulo SP, Brazil
| | - Daniele de Paula Faria
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Radiologia e Oncologia, Laboratório de Medicina Nuclear (LIM/43), São Paulo SP, Brazil
| | - Paula Squarzoni
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Psiquiatria, São Paulo SP, Brazil
| | - Geraldo Busatto Filho
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Psiquiatria, São Paulo SP, Brazil
| | - Carlos Alberto Buchpiguel
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Departamento de Radiologia e Oncologia, Laboratório de Medicina Nuclear (LIM/43), São Paulo SP, Brazil
| | - Ricardo Nitrini
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| | - Sonia Maria Dozzi Brucki
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Neurologia, Grupo de Neurologia Cognitiva e do Comportamento, São Paulo SP, Brazil
| |
Collapse
|
16
|
Tsuchida A, Goubet M, Boutinaud P, Astafeva I, Nozais V, Hervé PY, Tourdias T, Debette S, Joliot M. SHIVA-CMB: a deep-learning-based robust cerebral microbleed segmentation tool trained on multi-source T2*GRE- and susceptibility-weighted MRI. Sci Rep 2024; 14:30901. [PMID: 39730628 DOI: 10.1038/s41598-024-81870-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 11/29/2024] [Indexed: 12/29/2024] Open
Abstract
Cerebral microbleeds (CMB) represent a feature of cerebral small vessel disease (cSVD), a prominent vascular contributor to age-related cognitive decline, dementia, and stroke. They are visible as spherical hypointense signals on T2*- or susceptibility-weighted magnetic resonance imaging (MRI) sequences. An increasing number of automated CMB detection methods being proposed are based on supervised deep learning (DL). Yet, the lack of open sharing of pre-trained models hampers the practical application and evaluation of these methods beyond specific data sources used in each study. Here, we present the SHIVA-CMB detector, a 3D Unet-based tool trained on 450 scans taken from seven acquisitions in six different cohort studies that included both T2*- and susceptibility-weighted MRI. In a held-out test set of 96 scans, it achieved the sensitivity, precision, and F1 (or Dice similarity coefficient) score of 0.67, 0.82, and 0.74, with less than one false positive detection per image (FPavg = 0.6) and per CMB (FPcmb = 0.15). It achieved a similar level of performance in a separate, evaluation-only dataset with acquisitions never seen during the training (0.67, 0.91, 0.77, 0.5, 0.07 for the sensitivity, precision, F1 score, FPavg, and FPcmb). Further demonstrating its generalizability, it showed a high correlation (Pearson's R = 0.89, p < 0.0001) with a visual count by expert raters in another independent set of 1992 T2*-weighted scans from a large, multi-center cohort study. Importantly, we publicly share both the pipeline ( https://github.com/pboutinaud/SHiVAi/ ) and pre-trained models ( https://github.com/pboutinaud/SHIVA-CMB/ ) to the research community to promote the active application and evaluation of our tool. We believe this effort will help accelerate research on the pathophysiology and functional consequences of CMB by enabling rapid characterization of CMB in large-scale studies.
Collapse
Affiliation(s)
- Ami Tsuchida
- GIN, IMN-UMR5293, CEA, CNRS, Université de Bordeaux, Bordeaux, France
- BPH-U1219, INSERM, Université de Bordeaux, Bordeaux, France
| | | | | | - Iana Astafeva
- GIN, IMN-UMR5293, CEA, CNRS, Université de Bordeaux, Bordeaux, France
- BPH-U1219, INSERM, Université de Bordeaux, Bordeaux, France
| | | | | | - Thomas Tourdias
- Neuroimagerie diagnostique et thérapeutique, CHU de Bordeaux, Bordeaux, France
- Neurocentre Magendie-U1219, INSERM, Université de Bordeaux, Bordeaux, France
| | | | - Marc Joliot
- GIN, IMN-UMR5293, CEA, CNRS, Université de Bordeaux, Bordeaux, France.
| |
Collapse
|
17
|
Zhu Y, Wang F, Xia Y, Wang L, Lin H, Zhong T, Wang X. Research progress on astrocyte-derived extracellular vesicles in the pathogenesis and treatment of neurodegenerative diseases. Rev Neurosci 2024; 35:855-875. [PMID: 38889403 DOI: 10.1515/revneuro-2024-0043] [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: 03/26/2024] [Accepted: 05/24/2024] [Indexed: 06/20/2024]
Abstract
Neurodegenerative disorders, including Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD), pose significant global health risks and represent a substantial public health concern in the contemporary era. A primary factor in the pathophysiology of these disorders is aberrant accumulation and aggregation of pathogenic proteins within the brain and spinal cord. Recent investigations have identified extracellular vesicles (EVs) in the central nervous system (CNS) as potential carriers for intercellular transport of misfolded proteins associated with neurodegenerative diseases. EVs are involved in pathological processes that contribute to various brain disorders including neurodegenerative disorders. Proteins linked to neurodegenerative disorders are secreted and distributed from cell to cell via EVs, serving as a mechanism for direct intercellular communication through the transfer of biomolecules. Astrocytes, as active participants in CNS intercellular communication, release astrocyte-derived extracellular vesicles (ADEVs) that are capable of interacting with diverse target cells. This review primarily focuses on the involvement of ADEVs in the development of neurological disorders and explores their potential dual roles - both advantageous and disadvantageous in the context of neurological disorders. Furthermore, this review examines the current studies investigating ADEVs as potential biomarkers for the diagnosis and treatment of neurodegenerative diseases. The prospects and challenges associated with the application of ADEVs in clinical settings were also comprehensively reviewed.
Collapse
Affiliation(s)
- Yifan Zhu
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, 341000, Jiangxi, China
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Fangsheng Wang
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Yu Xia
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Lijuan Wang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, 341000, Jiangxi, China
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Haihong Lin
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, 341000, Jiangxi, China
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Tianyu Zhong
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, 341000, Jiangxi, China
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Xiaoling Wang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, 341000, Jiangxi, China
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| |
Collapse
|
18
|
Song Z, Li H, Zhang Y, Zhu C, Jiang M, Song L, Wang Y, Ouyang M, Hu F, Zheng Q. s 2MRI-ADNet: an interpretable deep learning framework integrating Euclidean-graph representations of Alzheimer's disease solely from structural MRI. MAGMA (NEW YORK, N.Y.) 2024; 37:845-857. [PMID: 38869733 DOI: 10.1007/s10334-024-01178-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/19/2024] [Accepted: 06/04/2024] [Indexed: 06/14/2024]
Abstract
OBJECTIVE To establish a multi-dimensional representation solely on structural MRI (sMRI) for early diagnosis of AD. METHODS A total of 3377 participants' sMRI from four independent databases were retrospectively identified to construct an interpretable deep learning model that integrated multi-dimensional representations of AD solely on sMRI (called s2MRI-ADNet) by a dual-channel learning strategy of gray matter volume (GMV) from Euclidean space and the regional radiomics similarity network (R2SN) from graph space. Specifically, the GMV feature map learning channel (called GMV-Channel) was to take into consideration spatial information of both long-range spatial relations and detailed localization information, while the node feature and connectivity strength learning channel (called NFCS-Channel) was to characterize the graph-structured R2SN network by a separable learning strategy. RESULTS The s2MRI-ADNet achieved a superior classification accuracy of 92.1% and 91.4% under intra-database and inter-database cross-validation. The GMV-Channel and NFCS-Channel captured complementary group-discriminative brain regions, revealing a complementary interpretation of the multi-dimensional representation of brain structure in Euclidean and graph spaces respectively. Besides, the generalizable and reproducible interpretation of the multi-dimensional representation in capturing complementary group-discriminative brain regions revealed a significant correlation between the four independent databases (p < 0.05). Significant associations (p < 0.05) between attention scores and brain abnormality, between classification scores and clinical measure of cognitive ability, CSF biomarker, metabolism, and genetic risk score also provided solid neurobiological interpretation. CONCLUSION The s2MRI-ADNet solely on sMRI could leverage the complementary multi-dimensional representations of AD in Euclidean and graph spaces, and achieved superior performance in the early diagnosis of AD, facilitating its potential in both clinical translation and popularization.
Collapse
Affiliation(s)
- Zhiwei Song
- School of Computer and Control Engineering, Yantai University, Yantai, 264005, China
| | - Honglun Li
- Department of Radiology, Yantai Yuhuangding Hospital Affiliated with Qingdao University Medical College, Yantai, 264099, China
| | - Yiyu Zhang
- School of Computer and Control Engineering, Yantai University, Yantai, 264005, China
| | - Chuanzhen Zhu
- School of Computer and Control Engineering, Yantai University, Yantai, 264005, China
| | - Minbo Jiang
- School of Computer and Control Engineering, Yantai University, Yantai, 264005, China
| | - Limei Song
- School of Medical Imaging, Weifang Medical University, Weifang, 261000, China
| | - Yi Wang
- School of Computer and Control Engineering, Yantai University, Yantai, 264005, China
- Key Laboratory of Medical Imaging and Artificial Intelligence of Hunan Province, Xiangnan University, Chenzhou, 423000, Hunan, China
| | - Minhui Ouyang
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Fang Hu
- Key Laboratory of Medical Imaging and Artificial Intelligence of Hunan Province, Xiangnan University, Chenzhou, 423000, Hunan, China
| | - Qiang Zheng
- School of Computer and Control Engineering, Yantai University, Yantai, 264005, China.
| |
Collapse
|
19
|
Huang X, Fowler C, Li Y, Li QX, Sun J, Pan Y, Jin L, Perez KA, Dubois C, Lim YY, Drysdale C, Rumble RL, Chinnery HR, Rowe CC, Martins RN, Maruff P, Doecke JD, Lin Y, Belaidi AA, Barnham KJ, Masters CL, Gu BJ. Clearance and transport of amyloid β by peripheral monocytes correlate with Alzheimer's disease progression. Nat Commun 2024; 15:7998. [PMID: 39266542 PMCID: PMC11393069 DOI: 10.1038/s41467-024-52396-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 09/02/2024] [Indexed: 09/14/2024] Open
Abstract
Impaired clearance of amyloid β (Aβ) in late-onset Alzheimer's disease (AD) affects disease progression. The role of peripheral monocytes in Aβ clearance from the central nervous system (CNS) is unclear. We use a flow cytometry assay to identify Aβ-binding monocytes in blood, validated by confocal microscopy, Western blotting, and mass spectrometry. Flow cytometry immunophenotyping and correlation with AD biomarkers are studied in 150 participants from the AIBL study. We also examine monocytes in human cerebrospinal fluid (CSF) and their migration in an APP/PS1 mouse model. The assay reveals macrophage-like Aβ-binding monocytes with high phagocytic potential in both the periphery and CNS. We find lower surface Aβ levels in mild cognitive impairment (MCI) and AD-dementia patients compared to cognitively unimpaired individuals. Monocyte infiltration from blood to CSF and migration from CNS to peripheral lymph nodes and blood are observed. Here we show that Aβ-binding monocytes may play a role in CNS Aβ clearance, suggesting their potential as a biomarker for AD diagnosis and monitoring.
Collapse
Affiliation(s)
- Xin Huang
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
- The Innate Phagocytosis Laboratory, Level 11, Melbourne, Victoria, Australia
| | - Chris Fowler
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Yihan Li
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Qiao-Xin Li
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
- National Dementia Diagnostics Laboratory, The University of Melbourne, Parkville, VIC, Australia
| | - Jiaqi Sun
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Yijun Pan
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Liang Jin
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Keyla A Perez
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Céline Dubois
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Yen Y Lim
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, VIC, Australia
| | - Candace Drysdale
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Rebecca L Rumble
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Holly R Chinnery
- Optometry and Vision Sciences, The University of Melbourne, Parkville, Victoria, Australia
- Lions Eye Institute, Perth, Western Australia, Australia
- Optometry, School of Allied Health, The University of Western Australia, Perth, Australia
| | - Christopher C Rowe
- Department of Nuclear Medicine and Center for PET, Austin Health, Heidelberg, VIC, Australia
| | - Ralph N Martins
- Center of Excellence for Alzheimer's Disease Research and Care, Edith Cowan University, Joondalup, WA, Australia
| | - Paul Maruff
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
- Cogstate Ltd., Melbourne, VIC, Australia
| | - James D Doecke
- Health and Biosecurity, Australian E-Health Research Center, CSIRO, Brisbane, QLD, Australia
| | - Yong Lin
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Abdel A Belaidi
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Kevin J Barnham
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Colin L Masters
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia.
| | - Ben J Gu
- The Florey Institute, The University of Melbourne, Parkville, VIC, Australia.
- The Innate Phagocytosis Laboratory, Level 11, Melbourne, Victoria, Australia.
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
20
|
Ma LY, Song JH, Gao PY, Ou YN, Fu Y, Huang LY, Wang ZT, Zhang DD, Cui RP, Mi YC, Tan L. Amyloid pathology mediates the associations between plasma fibrinogen and cognition in non-demented adults. J Neurochem 2024; 168:2532-2542. [PMID: 38533619 DOI: 10.1111/jnc.16105] [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: 01/29/2024] [Revised: 03/08/2024] [Accepted: 03/13/2024] [Indexed: 03/28/2024]
Abstract
Though previous studies revealed the potential associations of elevated levels of plasma fibrinogen with dementia, there is still limited understanding regarding the influence of Alzheimer's disease (AD) biomarkers on these associations. We sought to investigate the interrelationships among fibrinogen, cerebrospinal fluid (CSF) AD biomarkers, and cognition in non-demented adults. We included 1996 non-demented adults from the Chinese Alzheimer's Biomarker and LifestylE (CABLE) study and 337 from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database. The associations of fibrinogen with AD biomarkers and cognition were explored using multiple linear regression models. The mediation analyses with 10 000 bootstrapped iterations were conducted to explore the mediating effects of AD biomarkers on cognition. In addition, interaction analyses and subgroup analyses were conducted to assess the influence of covariates on the relationships between fibrinogen and AD biomarkers. Participants exhibiting low Aβ42 were designated as A+, while those demonstrating high phosphorylated tau (P-tau) and total tau (Tau) were labeled as T+ and N+, respectively. Individuals with normal measures of Aβ42 and P-tau were categorized as the A-T- group, and those with abnormal levels of both Aβ42 and P-tau were grouped under A+T+. Fibrinogen was higher in the A+ subgroup compared to that in the A- subgroup (p = 0.026). Fibrinogen was higher in the A+T+ subgroup compared to that in the A-T- subgroup (p = 0.011). Higher fibrinogen was associated with worse cognition and Aβ pathology (all p < 0.05). Additionally, the associations between fibrinogen and cognition were partially mediated by Aβ pathology (mediation proportion range 8%-28%). Interaction analyses and subgroup analyses showed that age and ApoE ε4 affect the relationships between fibrinogen and Aβ pathology. Fibrinogen was associated with both cognition and Aβ pathology. Aβ pathology may be a critical mediator for impacts of fibrinogen on cognition.
Collapse
Affiliation(s)
- Li-Yun Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jing-Hui Song
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Pei-Yang Gao
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yan Fu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Liang-Yu Huang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Zuo-Teng Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Dan-Dan Zhang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Rui-Ping Cui
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yin-Chu Mi
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| |
Collapse
|
21
|
Arenaza‐Urquijo EM, Boyle R, Casaletto K, Anstey KJ, Vila‐Castelar C, Colverson A, Palpatzis E, Eissman JM, Kheng Siang Ng T, Raghavan S, Akinci M, Vonk JMJ, Machado LS, Zanwar PP, Shrestha HL, Wagner M, Tamburin S, Sohrabi HR, Loi S, Bartrés‐Faz D, Dubal DB, Vemuri P, Okonkwo O, Hohman TJ, Ewers M, Buckley RF, for the Reserve, Resilience and Protective Factors Professional Interest Area, Sex and Gender Professional Interest area and the ADDRESS! Special Interest Group. Sex and gender differences in cognitive resilience to aging and Alzheimer's disease. Alzheimers Dement 2024; 20:5695-5719. [PMID: 38967222 PMCID: PMC11350140 DOI: 10.1002/alz.13844] [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: 11/08/2023] [Revised: 03/08/2024] [Accepted: 03/21/2024] [Indexed: 07/06/2024]
Abstract
Sex and gender-biological and social constructs-significantly impact the prevalence of protective and risk factors, influencing the burden of Alzheimer's disease (AD; amyloid beta and tau) and other pathologies (e.g., cerebrovascular disease) which ultimately shape cognitive trajectories. Understanding the interplay of these factors is central to understanding resilience and resistance mechanisms explaining maintained cognitive function and reduced pathology accumulation in aging and AD. In this narrative review, the ADDRESS! Special Interest Group (Alzheimer's Association) adopted a multidisciplinary approach to provide the foundations and recommendations for future research into sex- and gender-specific drivers of resilience, including a sex/gender-oriented review of risk factors, genetics, AD and non-AD pathologies, brain structure and function, and animal research. We urge the field to adopt a sex/gender-aware approach to resilience to advance our understanding of the intricate interplay of biological and social determinants and consider sex/gender-specific resilience throughout disease stages. HIGHLIGHTS: Sex differences in resilience to cognitive decline vary by age and cognitive status. Initial evidence supports sex-specific distinctions in brain pathology. Findings suggest sex differences in the impact of pathology on cognition. There is a sex-specific change in resilience in the transition to clinical stages. Gender and sex factors warrant study: modifiable, immune, inflammatory, and vascular.
Collapse
Affiliation(s)
- Eider M. Arenaza‐Urquijo
- Environment and Health Over the Life Course Programme, Climate, Air Pollution, Nature and Urban Health ProgrammeBarcelona Institute for Global Health (ISGlobal)BarcelonaSpain
- University of Pompeu FabraBarcelonaBarcelonaSpain
| | - Rory Boyle
- Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Kaitlin Casaletto
- Department of NeurologyMemory and Aging CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Kaarin J. Anstey
- University of New South Wales Ageing Futures InstituteSydneyNew South WalesAustralia
- Neuroscience Research AustraliaSydneyNew South WalesAustralia
- School of Psychology, University of New South WalesSidneyNew South WalesAustralia
| | | | - Aaron Colverson
- University of Florida Center for Arts in Medicine Interdisciplinary Research LabUniversity of Florida, Center of Arts in MedicineGainesvilleFloridaUSA
| | - Eleni Palpatzis
- Environment and Health Over the Life Course Programme, Climate, Air Pollution, Nature and Urban Health ProgrammeBarcelona Institute for Global Health (ISGlobal)BarcelonaSpain
- University of Pompeu FabraBarcelonaBarcelonaSpain
| | - Jaclyn M. Eissman
- Vanderbilt Memory and Alzheimer's Center, Department of NeurologyVanderbilt University Medical CenterNashvilleTennesseeUSA
- Vanderbilt Genetics InstituteVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Ted Kheng Siang Ng
- Rush Institute for Healthy Aging and Department of Internal MedicineRush University Medical CenterChicagoIllinoisUSA
| | | | - Muge Akinci
- Environment and Health Over the Life Course Programme, Climate, Air Pollution, Nature and Urban Health ProgrammeBarcelona Institute for Global Health (ISGlobal)BarcelonaSpain
- University of Pompeu FabraBarcelonaBarcelonaSpain
| | - Jet M. J. Vonk
- Department of NeurologyMemory and Aging CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Luiza S. Machado
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal Do Rio Grande Do Sul, FarroupilhaPorto AlegreBrazil
| | - Preeti P. Zanwar
- Jefferson College of Population Health, Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
- The Network on Life Course and Health Dynamics and Disparities, University of Southern CaliforniaLos AngelesCaliforniaUSA
| | | | - Maude Wagner
- Rush Alzheimer's Disease Center, Rush University Medical CenterChicagoIllinoisUSA
| | - Stefano Tamburin
- Department of Neurosciences, Biomedicine and Movement SciencesUniversity of VeronaVeronaItaly
| | - Hamid R. Sohrabi
- Centre for Healthy AgeingHealth Future InstituteMurdoch UniversityMurdochWestern AustraliaAustralia
- School of Psychology, Murdoch UniversityMurdochWestern AustraliaAustralia
| | - Samantha Loi
- Neuropsychiatry Centre, Royal Melbourne HospitalParkvilleVictoriaAustralia
- Department of PsychiatryUniversity of MelbourneParkvilleVictoriaAustralia
| | - David Bartrés‐Faz
- Department of MedicineFaculty of Medicine and Health Sciences & Institut de NeurociènciesUniversity of BarcelonaBarcelonaBarcelonaSpain
- Institut d'Investigacions Biomèdiques (IDIBAPS)BarcelonaBarcelonaSpain
- Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la Universitat Autónoma de BarcelonaBadalonaBarcelonaSpain
| | - Dena B. Dubal
- Department of Neurology and Weill Institute of NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Biomedical and Neurosciences Graduate ProgramsUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | | | - Ozioma Okonkwo
- Alzheimer's Disease Research Center and Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Timothy J. Hohman
- Vanderbilt Memory and Alzheimer's Center, Department of NeurologyVanderbilt University Medical CenterNashvilleTennesseeUSA
- Vanderbilt Genetics InstituteVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Michael Ewers
- Institute for Stroke and Dementia ResearchKlinikum der Universität MünchenLudwig Maximilians Universität (LMU)MunichGermany
- German Center for Neurodegenerative Diseases (DZNE, Munich)MunichGermany
| | - Rachel F. Buckley
- Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | | |
Collapse
|
22
|
Reitz NL, Nunes PT, Savage LM. Exercise leads to sex-specific recovery of behavior and pathological AD markers following adolescent ethanol exposure in the TgF344-AD model. Front Behav Neurosci 2024; 18:1448691. [PMID: 39148897 PMCID: PMC11324591 DOI: 10.3389/fnbeh.2024.1448691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 07/12/2024] [Indexed: 08/17/2024] Open
Abstract
Introduction Human epidemiological studies suggest that heavy alcohol consumption may lead to earlier onset of Alzheimer's Disease (AD), especially in individuals with a genetic predisposition for AD. Alcohol-related brain damage (ARBD) during a critical developmental timepoint, such as adolescence, interacts with AD-related pathologies to accelerate disease progression later in life. The current study investigates if voluntary exercise in mid-adulthood can recover memory deficits caused by the interactions between adolescence ethanol exposure and AD-transgenes. Methods Male and female TgF344-AD and wildtype F344 rats were exposed to an intragastric gavage of water (control) or 5 g/kg of 20% ethanol (adolescent intermittent ethanol; AIE) for a 2 day on/off schedule throughout adolescence (PD27-57). At 6 months old, rats either remained in their home cage (stationary) or were placed in a voluntary wheel running apparatus for 4 weeks and then underwent several behavioral tests. The number of cholinergic neurons in the basal forebrain and measure of neurogenesis in the hippocampus were assessed. Results Voluntary wheel running recovers spatial working memory deficits selectively in female TgF344-AD rats exposed to AIE and improves pattern separation impairment seen in control TgF344-AD female rats. There were sex-dependent effects on brain pathology: Exercise improves the integration of recently born neurons in AIE-exposed TgF344-AD female rats. Exercise led to a decrease in amyloid burden in the hippocampus and entorhinal cortex, but only in male AIE-exposed TgF344-AD rats. Although the number of basal forebrain cholinergic neurons was not affected by AD-transgenes in either sex, AIE did reduce the number of basal forebrain cholinergic neurons in female rats. Discussion These data provide support that even after symptom onset, AIE and AD related cognitive decline and associated neuropathologies can be rescued with exercise in unique sex-specific ways.
Collapse
Affiliation(s)
| | | | - Lisa M. Savage
- Department of Psychology, Binghamton University, State University of New York, Binghamton, NY, United States
| |
Collapse
|
23
|
Chattopadhyay T, Ozarkar SS, Buwa K, Joshy NA, Komandur D, Naik J, Thomopoulos SI, Ver Steeg G, Ambite JL, Thompson PM. Comparison of deep learning architectures for predicting amyloid positivity in Alzheimer's disease, mild cognitive impairment, and healthy aging, from T1-weighted brain structural MRI. Front Neurosci 2024; 18:1387196. [PMID: 39015378 PMCID: PMC11250587 DOI: 10.3389/fnins.2024.1387196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 06/14/2024] [Indexed: 07/18/2024] Open
Abstract
Abnormal β-amyloid (Aβ) accumulation in the brain is an early indicator of Alzheimer's disease (AD) and is typically assessed through invasive procedures such as PET (positron emission tomography) or CSF (cerebrospinal fluid) assays. As new anti-Alzheimer's treatments can now successfully target amyloid pathology, there is a growing interest in predicting Aβ positivity (Aβ+) from less invasive, more widely available types of brain scans, such as T1-weighted (T1w) MRI. Here we compare multiple approaches to infer Aβ + from standard anatomical MRI: (1) classical machine learning algorithms, including logistic regression, XGBoost, and shallow artificial neural networks, (2) deep learning models based on 2D and 3D convolutional neural networks (CNNs), (3) a hybrid ANN-CNN, combining the strengths of shallow and deep neural networks, (4) transfer learning models based on CNNs, and (5) 3D Vision Transformers. All models were trained on paired MRI/PET data from 1,847 elderly participants (mean age: 75.1 yrs. ± 7.6SD; 863 females/984 males; 661 healthy controls, 889 with mild cognitive impairment (MCI), and 297 with Dementia), scanned as part of the Alzheimer's Disease Neuroimaging Initiative. We evaluated each model's balanced accuracy and F1 scores. While further tests on more diverse data are warranted, deep learning models trained on standard MRI showed promise for estimating Aβ + status, at least in people with MCI. This may offer a potential screening option before resorting to more invasive procedures.
Collapse
Affiliation(s)
- Tamoghna Chattopadhyay
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, United States
| | - Saket S. Ozarkar
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, United States
| | - Ketaki Buwa
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, United States
| | - Neha Ann Joshy
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, United States
| | - Dheeraj Komandur
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, United States
| | - Jayati Naik
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, United States
| | - Sophia I. Thomopoulos
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, United States
| | | | - Jose Luis Ambite
- Information Sciences Institute, University of Southern California, Marina del Rey, CA, United States
| | - Paul M. Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, CA, United States
| |
Collapse
|
24
|
Strobel J, Yousefzadeh-Nowshahr E, Deininger K, Bohn KP, von Arnim CAF, Otto M, Solbach C, Anderl-Straub S, Polivka D, Fissler P, Glatting G, Riepe MW, Higuchi M, Beer AJ, Ludolph A, Winter G. Exploratory Tau PET/CT with [11C]PBB3 in Patients with Suspected Alzheimer's Disease and Frontotemporal Lobar Degeneration: A Pilot Study on Correlation with PET Imaging and Cerebrospinal Fluid Biomarkers. Biomedicines 2024; 12:1460. [PMID: 39062033 PMCID: PMC11274645 DOI: 10.3390/biomedicines12071460] [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: 05/08/2024] [Revised: 06/13/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Accurately diagnosing Alzheimer's disease (AD) and frontotemporal lobar degeneration (FTLD) is challenging due to overlapping symptoms and limitations of current imaging methods. This study investigates the use of [11C]PBB3 PET/CT imaging to visualize tau pathology and improve diagnostic accuracy. Given diagnostic challenges with symptoms and conventional imaging, [11C]PBB3 PET/CT's potential to enhance accuracy was investigated by correlating tau pathology with cerebrospinal fluid (CSF) biomarkers, positron emission tomography (PET), computed tomography (CT), amyloid-beta, and Mini-Mental State Examination (MMSE). We conducted [11C]PBB3 PET/CT imaging on 24 patients with suspected AD or FTLD, alongside [11C]PiB PET/CT (13 patients) and [18F]FDG PET/CT (15 patients). Visual and quantitative assessments of [11C]PBB3 uptake using standardized uptake value ratios (SUV-Rs) and correlation analyses with clinical assessments were performed. The scans revealed distinct tau accumulation patterns; 13 patients had no or faint uptake (PBB3-negative) and 11 had moderate to pronounced uptake (PBB3-positive). Significant inverse correlations were found between [11C]PBB3 SUV-Rs and MMSE scores, but not with CSF-tau or CSF-amyloid-beta levels. Here, we show that [11C]PBB3 PET/CT imaging can reveal distinct tau accumulation patterns and correlate these with cognitive impairment in neurodegenerative diseases. Our study demonstrates the potential of [11C]PBB3-PET imaging for visualizing tau pathology and assessing disease severity, offering a promising tool for enhancing diagnostic accuracy in AD and FTLD. Further research is essential to validate these findings and refine the use of tau-specific PET imaging in clinical practice, ultimately improving patient care and treatment outcomes.
Collapse
Affiliation(s)
- Joachim Strobel
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | | | - Katharina Deininger
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Karl Peter Bohn
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | | | - Markus Otto
- Department of Neurology, Halle University, 06120 Halle, Germany
| | - Christoph Solbach
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | | | - Dörte Polivka
- Department of Neurology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Patrick Fissler
- Psychiatric Services Thurgau (Academic Teaching Hospital of the University of Konstanz), 8596 Münsterlingen, Switzerland
| | - Gerhard Glatting
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Matthias W. Riepe
- Department of Psychiatry and Psychotherapy II, Ulm University, 89075 Ulm, Germany
| | - Makoto Higuchi
- National Institute of Radiological Sciences, Chiba 263-8555, Japan
| | - Ambros J. Beer
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| | - Albert Ludolph
- Department of Neurology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Gordon Winter
- Department of Nuclear Medicine, Ulm University Medical Center, 89081 Ulm, Germany
| |
Collapse
|
25
|
Ishiki K, Yamashita K, Watanabe S, Miura M, Kawahira J, Arimatsu Y, Kawasaki K, Iwanaga S, Sato T. The appropriate sample-handling procedure for measuring the plasma β-amyloid level using a fully automated immunoassay. Sci Rep 2024; 14:14266. [PMID: 38902510 PMCID: PMC11190145 DOI: 10.1038/s41598-024-65264-1] [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: 02/22/2024] [Accepted: 06/18/2024] [Indexed: 06/22/2024] Open
Abstract
Plasma β-amyloid (Aβ) assays are a promising tool for Alzheimer's disease diagnosis in clinical practice. To obtain reliable results, establishing an appropriate sample-handling procedure for each analytical platform is warranted. This study proposes an appropriate sample-handling procedure using HISCL analyzer by elucidating the individual/combined effects of pre-analytical parameters on plasma Aβ42/Aβ40 levels. We investigated the effects of various pre-analytical parameters, including storage times for whole blood, plasma, and freezing conditions, on plasma Aβ42/Aβ40 levels, and confirmed if these values met the acceptable criteria. Plasma Aβ42/Aβ40 levels were acceptable in all conditions. We determined our protocol by confirming that plasma Aβ42/Aβ40 levels remained acceptable when combining pre-analytical parameters. We established an appropriate sample-handling protocol that ensures reliable measurement of plasma Aβ42/Aβ40 levels using HISCL analyzer. We believe the Aβ assay, with our protocol, shows promise for aiding AD diagnosis in clinical settings.
Collapse
Affiliation(s)
- Kengo Ishiki
- Central Research Laboratories, Sysmex Corporation, 4-4-4 Takatsukadai, Nishi-Ku, Kobe, 651-2271, Japan
| | - Kazuto Yamashita
- Central Research Laboratories, Sysmex Corporation, 4-4-4 Takatsukadai, Nishi-Ku, Kobe, 651-2271, Japan
| | - Shunsuke Watanabe
- Central Research Laboratories, Sysmex Corporation, 4-4-4 Takatsukadai, Nishi-Ku, Kobe, 651-2271, Japan
| | - Masahiro Miura
- Central Research Laboratories, Sysmex Corporation, 4-4-4 Takatsukadai, Nishi-Ku, Kobe, 651-2271, Japan.
| | - Junko Kawahira
- Reagent Engineering, Sysmex Corporation, 4-4-4 Takatsukadai, Nishi-Ku, Kobe, 651-2271, Japan
| | - Yuji Arimatsu
- Reagent Engineering, Sysmex Corporation, 4-4-4 Takatsukadai, Nishi-Ku, Kobe, 651-2271, Japan
| | - Kana Kawasaki
- Central Research Laboratories, Sysmex Corporation, 4-4-4 Takatsukadai, Nishi-Ku, Kobe, 651-2271, Japan
| | - Shigeki Iwanaga
- Central Research Laboratories, Sysmex Corporation, 4-4-4 Takatsukadai, Nishi-Ku, Kobe, 651-2271, Japan
| | - Toshiyuki Sato
- Central Research Laboratories, Sysmex Corporation, 4-4-4 Takatsukadai, Nishi-Ku, Kobe, 651-2271, Japan
| |
Collapse
|
26
|
Cody KA, Langhough RE, Zammit MD, Clark L, Chin N, Christian BT, Betthauser TJ, Johnson SC. Characterizing brain tau and cognitive decline along the amyloid timeline in Alzheimer's disease. Brain 2024; 147:2144-2157. [PMID: 38667631 PMCID: PMC11146417 DOI: 10.1093/brain/awae116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/23/2024] [Accepted: 03/24/2024] [Indexed: 06/04/2024] Open
Abstract
Recent longitudinal PET imaging studies have established methods to estimate the age at which amyloid becomes abnormal at the level of the individual. Here we recontextualized amyloid levels into the temporal domain to better understand the downstream Alzheimer's disease processes of tau neurofibrillary tangle (NFT) accumulation and cognitive decline. This cohort study included a total of 601 individuals from the Wisconsin Registry for Alzheimer's Prevention and Wisconsin Alzheimer's Disease Research Center that underwent amyloid and tau PET, longitudinal neuropsychological assessments and met clinical criteria for three clinical diagnosis groups: cognitively unimpaired (n = 537); mild cognitive impairment (n = 48); or dementia (n = 16). Cortical 11C-Pittsburgh compound B (PiB) distribution volume ratio (DVR) and sampled iterative local approximation were used to estimate amyloid positive (A+; global PiB DVR > 1.16 equivalent to 17.1 centiloids) onset age and years of A+ duration at tau PET (i.e. amyloid chronicity). Tau PET burden was quantified using 18F-MK-6240 standardized uptake value ratios (70-90 min, inferior cerebellar grey matter reference region). Whole-brain and region-specific approaches were used to examine tau PET binding along the amyloid timeline and across the Alzheimer's disease clinical continuum. Voxel-wise 18F-MK-6240 analyses revealed that with each decade of A+, the spatial extent of measurable tau spread (i.e. progressed) from regions associated with early to late NFT tau stages. Regional analyses indicated that tau burden in the entorhinal cortex was detectable, on average, within 10 years of A+ onset. Additionally, the entorhinal cortex was the region most sensitive to early amyloid pathology and clinical impairment in this predominantly preclinical sample. Among initially cognitively unimpaired (n = 472) individuals with longitudinal cognitive follow-up, mixed effects models showed significant linear and non-linear interactions of A+ duration and entorhinal tau on cognitive decline, suggesting a synergistic effect whereby greater A+ duration, together with a higher entorhinal tau burden, increases the likelihood of cognitive decline beyond their separable effects. Overall, the amyloid time framework enabled a spatiotemporal characterization of tau deposition patterns across the Alzheimer's disease continuum. This approach, which examined cross-sectional tau PET data along the amyloid timeline to make longitudinal disease course inferences, demonstrated that A+ duration explains a considerable amount of variability in the magnitude and topography of tau spread, which largely recapitulated NFT staging observed in human neuropathological studies. By anchoring disease progression to the onset of amyloid, this study provides a temporal disease context, which may help inform disease prognosis and timing windows for anti-amyloid therapies.
Collapse
Affiliation(s)
- Karly A Cody
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Rebecca E Langhough
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Matthew D Zammit
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin-Madison, Madison, WI 53792, USA
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Lindsay Clark
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Nathaniel Chin
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Bradley T Christian
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin-Madison, Madison, WI 53792, USA
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Tobey J Betthauser
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI 53792, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| |
Collapse
|
27
|
Ourry V, Binette AP, St-Onge F, Strikwerda-Brown C, Chagnot A, Poirier J, Breitner J, Arenaza-Urquijo EM, Rabin JS, Buckley R, Gonneaud J, Marchant NL, Villeneuve S. How Do Modifiable Risk Factors Affect Alzheimer's Disease Pathology or Mitigate Its Effect on Clinical Symptom Expression? Biol Psychiatry 2024; 95:1006-1019. [PMID: 37689129 DOI: 10.1016/j.biopsych.2023.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 08/11/2023] [Accepted: 09/03/2023] [Indexed: 09/11/2023]
Abstract
Epidemiological studies show that modifiable risk factors account for approximately 40% of the population variability in risk of developing dementia, including sporadic Alzheimer's disease (AD). Recent findings suggest that these factors may also modify disease trajectories of people with autosomal-dominant AD. With positron emission tomography imaging, it is now possible to study the disease many years before its clinical onset. Such studies can provide key knowledge regarding pathways for either the prevention of pathology or the postponement of its clinical expression. The former "resistance pathway" suggests that modifiable risk factors could affect amyloid and tau burden decades before the appearance of cognitive impairment. Alternatively, the resilience pathway suggests that modifiable risk factors may mitigate the symptomatic expression of AD pathology on cognition. These pathways are not mutually exclusive and may appear at different disease stages. Here, in a narrative review, we present neuroimaging evidence that supports both pathways in sporadic AD and autosomal-dominant AD. We then propose mechanisms for their protective effect. Among possible mechanisms, we examine neural and vascular mechanisms for the resistance pathway. We also describe brain maintenance and functional compensation as bases for the resilience pathway. Improved mechanistic understanding of both pathways may suggest new interventions.
Collapse
Affiliation(s)
- Valentin Ourry
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada.
| | - Alexa Pichet Binette
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada; Clinical Memory Research Unit, Department of Clinical Sciences, Lunds Universitet, Malmö, Sweden
| | - Frédéric St-Onge
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada; Integrated Program in Neuroscience, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Cherie Strikwerda-Brown
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada; School of Psychological Science, The University of Western Australia, Perth, Western Australia, Australia
| | - Audrey Chagnot
- UK Dementia Research Institute, Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Judes Poirier
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - John Breitner
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Eider M Arenaza-Urquijo
- Environment and Health over the Lifecourse Programme, Barcelona Institute for Global Health (ISGlobal), Barcelona, Spain; Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | - Jennifer S Rabin
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada; Harquail Centre for Neuromodulation, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada; Rehabilitation Sciences Institute, University of Toronto, Toronto, Ontario, Canada
| | - Rachel Buckley
- Melbourne School of Psychological Sciences University of Melbourne, Parkville, Victoria, Australia; Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts; Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Julie Gonneaud
- Normandie University, UNICAEN, INSERM, U1237, PhIND "Physiopathology and Imaging of Neurological Disorders," Institut Blood and Brain @ Caen-Normandie, GIP Cyceron, Caen, France
| | - Natalie L Marchant
- Division of Psychiatry, University College London, London, United Kingdom
| | - Sylvia Villeneuve
- Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada; McConnell Brain Imaging Center, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
28
|
Reuben DB, Kremen S, Maust DT. Dementia Prevention and Treatment: A Narrative Review. JAMA Intern Med 2024; 184:563-572. [PMID: 38436963 DOI: 10.1001/jamainternmed.2023.8522] [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] [Indexed: 03/05/2024]
Abstract
Importance Dementia affects 10% of those 65 years or older and 35% of those 90 years or older, often with profound cognitive, behavioral, and functional consequences. As the baby boomers and subsequent generations age, effective preventive and treatment strategies will assume increasing importance. Observations Preventive measures are aimed at modifiable risk factors, many of which have been identified. To date, no randomized clinical trial data conclusively confirm that interventions of any kind can prevent dementia. Nevertheless, addressing risk factors may have other health benefits and should be considered. Alzheimer disease can be treated with cholinesterase inhibitors, memantine, and antiamyloid immunomodulators, with the last modestly slowing cognitive and functional decline in people with mild cognitive impairment or mild dementia due to Alzheimer disease. Cholinesterase inhibitors and memantine may benefit persons with other types of dementia, including dementia with Lewy bodies, Parkinson disease dementia, vascular dementia, and dementia due to traumatic brain injury. Behavioral and psychological symptoms of dementia are best treated with nonpharmacologic management, including identifying and mitigating the underlying causes and individually tailored behavioral approaches. Psychotropic medications have minimal evidence of efficacy for treating these symptoms and are associated with increased mortality and clinically meaningful risks of falls and cognitive decline. Several emerging prevention and treatment strategies hold promise to improve dementia care in the future. Conclusions and Relevance Although current prevention and treatment approaches to dementia have been less than optimally successful, substantial investments in dementia research will undoubtedly provide new answers to reducing the burden of dementia worldwide.
Collapse
Affiliation(s)
- David B Reuben
- Multicampus Program in Geriatric Medicine and Gerontology, David Geffen School of Medicine, University of California, Los Angeles
| | - Sarah Kremen
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California
- Jona Goldrich Center for Alzheimer's and Memory Disorders, Cedars-Sinai Medical Center, Los Angeles, California
| | - Donovan T Maust
- Department of Psychiatry, University of Michigan, Ann Arbor
- Center for Clinical Management Research, VA Ann Arbor Healthcare System, Ann Arbor, Michigan
| |
Collapse
|
29
|
Aceves M, Granados J, Leandro AC, Peralta J, Glahn DC, Williams-Blangero S, Curran JE, Blangero J, Kumar S. Role of Neurocellular Endoplasmic Reticulum Stress Response in Alzheimer's Disease and Related Dementias Risk. Genes (Basel) 2024; 15:569. [PMID: 38790197 PMCID: PMC11121587 DOI: 10.3390/genes15050569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 04/24/2024] [Accepted: 04/26/2024] [Indexed: 05/26/2024] Open
Abstract
Currently, more than 55 million people around the world suffer from dementia, and Alzheimer's Disease and Related Dementias (ADRD) accounts for nearly 60-70% of all those cases. The spread of Alzheimer's Disease (AD) pathology and progressive neurodegeneration in the hippocampus and cerebral cortex is strongly correlated with cognitive decline in AD patients; however, the molecular underpinning of ADRD's causality is still unclear. Studies of postmortem AD brains and animal models of AD suggest that elevated endoplasmic reticulum (ER) stress may have a role in ADRD pathology through altered neurocellular homeostasis in brain regions associated with learning and memory. To study the ER stress-associated neurocellular response and its effects on neurocellular homeostasis and neurogenesis, we modeled an ER stress challenge using thapsigargin (TG), a specific inhibitor of sarco/endoplasmic reticulum Ca2+ ATPase (SERCA), in the induced pluripotent stem cell (iPSC)-derived neural stem cells (NSCs) of two individuals from our Mexican American Family Study (MAFS). High-content screening and transcriptomic analysis of the control and ER stress-challenged NSCs showed that the NSCs' ER stress response resulted in a significant decline in NSC self-renewal and an increase in apoptosis and cellular oxidative stress. A total of 2300 genes were significantly (moderated t statistics FDR-corrected p-value ≤ 0.05 and fold change absolute ≥ 2.0) differentially expressed (DE). The pathway enrichment and gene network analysis of DE genes suggests that all three unfolded protein response (UPR) pathways, protein kinase RNA-like ER kinase (PERK), activating transcription factor-6 (ATF-6), and inositol-requiring enzyme-1 (IRE1), were significantly activated and cooperatively regulated the NSCs' transcriptional response to ER stress. Our results show that IRE1/X-box binding protein 1 (XBP1) mediated transcriptional regulation of the E2F transcription factor 1 (E2F1) gene, and its downstream targets have a dominant role in inducing G1/S-phase cell cycle arrest in ER stress-challenged NSCs. The ER stress-challenged NSCs also showed the activation of C/EBP homologous protein (CHOP)-mediated apoptosis and the dysregulation of synaptic plasticity and neurotransmitter homeostasis-associated genes. Overall, our results suggest that the ER stress-associated attenuation of NSC self-renewal, increased apoptosis, and dysregulated synaptic plasticity and neurotransmitter homeostasis plausibly play a role in the causation of ADRD.
Collapse
Affiliation(s)
- Miriam Aceves
- Division of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, McAllen, TX 78504, USA; (M.A.); (J.G.)
| | - Jose Granados
- Division of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, McAllen, TX 78504, USA; (M.A.); (J.G.)
| | - Ana C. Leandro
- Division of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX 78520, USA; (A.C.L.); (J.P.); (S.W.-B.); (J.E.C.); (J.B.)
| | - Juan Peralta
- Division of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX 78520, USA; (A.C.L.); (J.P.); (S.W.-B.); (J.E.C.); (J.B.)
| | - David C. Glahn
- Department of Psychiatry, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA;
| | - Sarah Williams-Blangero
- Division of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX 78520, USA; (A.C.L.); (J.P.); (S.W.-B.); (J.E.C.); (J.B.)
| | - Joanne E. Curran
- Division of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX 78520, USA; (A.C.L.); (J.P.); (S.W.-B.); (J.E.C.); (J.B.)
| | - John Blangero
- Division of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX 78520, USA; (A.C.L.); (J.P.); (S.W.-B.); (J.E.C.); (J.B.)
| | - Satish Kumar
- Division of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, McAllen, TX 78504, USA; (M.A.); (J.G.)
| |
Collapse
|
30
|
Huszár Z, Engh MA, Pavlekovics M, Sato T, Steenkamp Y, Hanseeuw B, Terebessy T, Molnár Z, Hegyi P, Csukly G. Risk of conversion to mild cognitive impairment or dementia among subjects with amyloid and tau pathology: a systematic review and meta-analysis. Alzheimers Res Ther 2024; 16:81. [PMID: 38610055 PMCID: PMC11015617 DOI: 10.1186/s13195-024-01455-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 04/08/2024] [Indexed: 04/14/2024]
Abstract
BACKGROUND Measurement of beta-amyloid (Aβ) and phosphorylated tau (p-tau) levels offers the potential for early detection of neurocognitive impairment. Still, the probability of developing a clinical syndrome in the presence of these protein changes (A+ and T+) remains unclear. By performing a systematic review and meta-analysis, we investigated the risk of mild cognitive impairment (MCI) or dementia in the non-demented population with A+ and A- alone and in combination with T+ and T- as confirmed by PET or cerebrospinal fluid examination. METHODS A systematic search of prospective and retrospective studies investigating the association of Aβ and p-tau with cognitive decline was performed in three databases (MEDLINE via PubMed, EMBASE, and CENTRAL) on January 9, 2024. The risk of bias was assessed using the Cochrane QUIPS tool. Odds ratios (OR) and Hazard Ratios (HR) were pooled using a random-effects model. The effect of neurodegeneration was not studied due to its non-specific nature. RESULTS A total of 18,162 records were found, and at the end of the selection process, data from 36 cohorts were pooled (n= 7,793). Compared to the unexposed group, the odds ratio (OR) for conversion to dementia in A+ MCI patients was 5.18 [95% CI 3.93; 6.81]. In A+ CU subjects, the OR for conversion to MCI or dementia was 5.79 [95% CI 2.88; 11.64]. Cerebrospinal fluid Aβ42 or Aβ42/40 analysis and amyloid PET imaging showed consistent results. The OR for conversion in A+T+ MCI subjects (11.60 [95% CI 7.96; 16.91]) was significantly higher than in A+T- subjects (2.73 [95% CI 1.65; 4.52]). The OR for A-T+ MCI subjects was non-significant (1.47 [95% CI 0.55; 3.92]). CU subjects with A+T+ status had a significantly higher OR for conversion (13.46 [95% CI 3.69; 49.11]) than A+T- subjects (2.04 [95% CI 0.70; 5.97]). Meta-regression showed that the ORs for Aβ exposure decreased with age in MCI. (beta = -0.04 [95% CI -0.03 to -0.083]). CONCLUSIONS Identifying Aβ-positive individuals, irrespective of the measurement technique employed (CSF or PET), enables the detection of the most at-risk population before disease onset, or at least at a mild stage. The inclusion of tau status in addition to Aβ, especially in A+T+ cases, further refines the risk assessment. Notably, the higher odds ratio associated with Aβ decreases with age. TRIAL REGISTRATION The study was registered in PROSPERO (ID: CRD42021288100).
Collapse
Affiliation(s)
- Zsolt Huszár
- Centre for Translational Medicine, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
- Department of Psychiatry and Psychotherapy, Semmelweis University, Balassa utca 6, Budapest, 1083, Hungary
| | - Marie Anne Engh
- Centre for Translational Medicine, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
| | - Márk Pavlekovics
- Centre for Translational Medicine, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
- Department of Neurology, Jahn Ferenc Teaching Hospital, Köves utca 1, Budapest, 1204, Hungary
| | - Tomoya Sato
- Centre for Translational Medicine, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
| | - Yalea Steenkamp
- Centre for Translational Medicine, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
| | - Bernard Hanseeuw
- Department of Neurology and Institute of Neuroscience, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, 1200, Belgium
- Department of Radiology, Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02155, USA
| | - Tamás Terebessy
- Centre for Translational Medicine, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
| | - Zsolt Molnár
- Centre for Translational Medicine, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
- Department of Anesthesiology and Intensive Therapy, Semmelweis University, Üllői út 78/A, Budapest, Hungary
- Department of Anesthesiology and Intensive Therapy, Poznan University of Medical Sciences, 49 Przybyszewskiego St, Poznan, Poland
| | - Péter Hegyi
- Centre for Translational Medicine, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, Pécs, 7624, Hungary
- Institute of Pancreatic Diseases, Semmelweis University, Tömő 25-29, Budapest, 1083, Hungary
- Translational Pancreatology Research Group, Interdisciplinary Centre of Excellence for Research Development and Innovation University of Szeged, Budapesti 9, Szeged, 6728, Hungary
| | - Gábor Csukly
- Centre for Translational Medicine, Semmelweis University, Üllői út 26, Budapest, 1085, Hungary.
- Department of Psychiatry and Psychotherapy, Semmelweis University, Balassa utca 6, Budapest, 1083, Hungary.
| |
Collapse
|
31
|
de Vries LE, Huitinga I, Kessels HW, Swaab DF, Verhaagen J. The concept of resilience to Alzheimer's Disease: current definitions and cellular and molecular mechanisms. Mol Neurodegener 2024; 19:33. [PMID: 38589893 PMCID: PMC11003087 DOI: 10.1186/s13024-024-00719-7] [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: 08/23/2023] [Accepted: 03/20/2024] [Indexed: 04/10/2024] Open
Abstract
Some individuals are able to maintain their cognitive abilities despite the presence of significant Alzheimer's Disease (AD) neuropathological changes. This discrepancy between cognition and pathology has been labeled as resilience and has evolved into a widely debated concept. External factors such as cognitive stimulation are associated with resilience to AD, but the exact cellular and molecular underpinnings are not completely understood. In this review, we discuss the current definitions used in the field, highlight the translational approaches used to investigate resilience to AD and summarize the underlying cellular and molecular substrates of resilience that have been derived from human and animal studies, which have received more and more attention in the last few years. From these studies the picture emerges that resilient individuals are different from AD patients in terms of specific pathological species and their cellular reaction to AD pathology, which possibly helps to maintain cognition up to a certain tipping point. Studying these rare resilient individuals can be of great importance as it could pave the way to novel therapeutic avenues for AD.
Collapse
Affiliation(s)
- Luuk E de Vries
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, The Netherlands.
| | - Inge Huitinga
- Department of Neuroimmunology, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, The Netherlands
| | - Helmut W Kessels
- Swammerdam Institute for Life Sciences, Amsterdam Neuroscience, University of Amsterdam, 1098 XH, Amsterdam, the Netherlands
| | - Dick F Swaab
- Department of Neuropsychiatric Disorders, Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, Netherlands
| | - Joost Verhaagen
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, Institute of the Royal Netherlands Academy of Arts and Sciences, 1105 BA, Amsterdam, The Netherlands
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University, Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
| |
Collapse
|
32
|
Pivac LN, Brown BM, Sewell KR, Doecke JD, Villemagne VL, Doré V, Weinborn M, Sohrabi HR, Gardener SL, Bucks RS, Laws SM, Taddei K, Maruff P, Masters CL, Rowe C, Martins RN, Rainey‐Smith SR. Suboptimal self-reported sleep efficiency and duration are associated with faster accumulation of brain amyloid beta in cognitively unimpaired older adults. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12579. [PMID: 38651160 PMCID: PMC11033837 DOI: 10.1002/dad2.12579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 02/28/2024] [Accepted: 03/10/2024] [Indexed: 04/25/2024]
Abstract
INTRODUCTION This study investigated whether self-reported sleep quality is associated with brain amyloid beta (Aβ) accumulation. METHODS Linear mixed effect model analyses were conducted for 189 cognitively unimpaired (CU) older adults (mean ± standard deviation 74.0 ± 6.2; 53.2% female), with baseline self-reported sleep data, and positron emission tomography-determined brain Aβ measured over a minimum of three time points (range 33.3-72.7 months). Analyses included random slopes and intercepts, interaction for apolipoprotein E (APOE) ε4 allele status, and time, adjusting for sex and baseline age. RESULTS Sleep duration <6 hours, in APOE ε4 carriers, and sleep efficiency <65%, in the whole sample and APOE ε4 non-carriers, is associated with faster accumulation of brain Aβ. DISCUSSION These findings suggest a role for self-reported suboptimal sleep efficiency and duration in the accumulation of Alzheimer's disease (AD) neuropathology in CU individuals. Additionally, poor sleep efficiency represents a potential route via which individuals at lower genetic risk may progress to preclinical AD. Highlights In cognitively unimpaired older adults self-report sleep is associated with brain amyloid beta (Aβ) accumulation.Across sleep characteristics, this relationship differs by apolipoprotein E (APOE) genotype.Sleep duration <6 hours is associated with faster brain Aβ accumulation in APOE ε4 carriers.Sleep efficiency < 65% is associated with faster brain Aβ accumulation in APOE ε4 non-carriers.Personalized sleep interventions should be studied for potential to slow Aβ accumulation.
Collapse
Affiliation(s)
- Louise N. Pivac
- Centre for Healthy Ageing, Health Futures InstituteMurdoch UniversityMurdochWestern AustraliaAustralia
- Alzheimer's Research Australia, Sarich Neuroscience Research InstituteNedlandsWestern AustraliaAustralia
| | - Belinda M. Brown
- Centre for Healthy Ageing, Health Futures InstituteMurdoch UniversityMurdochWestern AustraliaAustralia
| | - Kelsey R. Sewell
- Centre for Healthy Ageing, Health Futures InstituteMurdoch UniversityMurdochWestern AustraliaAustralia
| | - James D. Doecke
- Australian E‐Health Research Centre, CSIROHerstonQueenslandAustralia
| | | | - Vincent Doré
- Australian E‐Health Research Centre, CSIROHerstonQueenslandAustralia
- Department of Molecular ImagingAustin HealthHeidelbergVictoriaAustralia
| | - Michael Weinborn
- School of Psychological ScienceUniversity of Western AustraliaPerthWestern AustraliaAustralia
| | - Hamid R. Sohrabi
- Centre for Healthy Ageing, Health Futures InstituteMurdoch UniversityMurdochWestern AustraliaAustralia
| | - Samantha L. Gardener
- School of Medical and Health SciencesEdith Cowan UniversityJoondalupWestern AustraliaAustralia
| | - Romola S. Bucks
- School of Psychological ScienceUniversity of Western AustraliaPerthWestern AustraliaAustralia
- School of Population and Global HealthUniversity of Western AustraliaPerthWestern AustraliaAustralia
| | - Simon M. Laws
- School of Medical and Health SciencesEdith Cowan UniversityJoondalupWestern AustraliaAustralia
- Centre for Precision HealthEdith Cowan UniversityJoondalupWestern AustraliaAustralia
- Collaborative Genomics and Translation GroupEdith Cowan UniversityJoondalupWestern AustraliaAustralia
- Curtin Medical SchoolCurtin UniversityBentleyWestern AustraliaAustralia
| | - Kevin Taddei
- School of Medical and Health SciencesEdith Cowan UniversityJoondalupWestern AustraliaAustralia
| | - Paul Maruff
- Cogstate Ltd., MelbourneMelbourneVictoriaAustralia
- The Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneMelbourneVictoriaAustralia
| | - Colin L. Masters
- The Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneMelbourneVictoriaAustralia
| | - Christopher Rowe
- Department of Molecular ImagingAustin HealthHeidelbergVictoriaAustralia
- The Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneMelbourneVictoriaAustralia
| | - Ralph N. Martins
- School of Medical and Health SciencesEdith Cowan UniversityJoondalupWestern AustraliaAustralia
- Department of Biomedical SciencesMacquarie UniversityMacquarie UniversitySydneyNew South WalesAustralia
| | - Stephanie R. Rainey‐Smith
- Centre for Healthy Ageing, Health Futures InstituteMurdoch UniversityMurdochWestern AustraliaAustralia
- Alzheimer's Research Australia, Sarich Neuroscience Research InstituteNedlandsWestern AustraliaAustralia
- School of Psychological ScienceUniversity of Western AustraliaPerthWestern AustraliaAustralia
- School of Medical and Health SciencesEdith Cowan UniversityJoondalupWestern AustraliaAustralia
| |
Collapse
|
33
|
Sun Z, Liu X, Pan D, Wang J. Unveiling the panorama and emerging boundaries in the field of aging biomarkers. Asian J Surg 2024; 47:2065-2066. [PMID: 38238139 DOI: 10.1016/j.asjsur.2024.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 01/05/2024] [Indexed: 04/04/2024] Open
Affiliation(s)
- Zhou Sun
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Xingzi Liu
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, 100034, China
| | - Dikang Pan
- Vascular Surgery Department, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Jingyu Wang
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, 100034, China.
| |
Collapse
|
34
|
Evans SA, Paitel ER, Bhasin R, Nielson KA. Genetic Risk for Alzheimer's Disease Alters Perceived Executive Dysfunction in Cognitively Healthy Middle-Aged and Older Adults. J Alzheimers Dis Rep 2024; 8:267-279. [PMID: 38405345 PMCID: PMC10894609 DOI: 10.3233/adr-230166] [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: 11/10/2023] [Accepted: 01/17/2024] [Indexed: 02/27/2024] Open
Abstract
Background Subjective cognitive complaints (SCC) may be an early indicator of future cognitive decline. However, findings comparing SCC and objective cognitive performance have varied, particularly in the memory domain. Even less well established is the relationship between subjective and objective complaints in non-amnestic domains, such as in executive functioning, despite evidence indicating very early changes in these domains. Moreover, particularly early changes in both amnestic and non-amnestic domains are apparent in those carrying the Apolipoprotein-E ɛ4 allele, a primary genetic risk for Alzheimer's disease (AD). Objective This study investigated the role of the ɛ4 allele in the consistency between subjective and objective executive functioning in 54 healthy, cognitively intact, middle-aged and older adults. Methods Participants (Mage = 64.07, SD = 9.27, range = 48-84; ɛ4+ = 18) completed the Frontal Systems Behavior Scale (FrSBe) Executive Dysfunction Scale (EXECDYS) to measure subjective executive functioning (SEF) and multiple executive functioning tasks, which were condensed into a single factor. Results After accounting for age, depression, and anxiety, objective executive functioning performance significantly predicted SEF. Importantly, ɛ4 moderated this effect. Specifically, those carrying the ɛ4 allele had significantly less accurate self-awareness of their executive functioning compared to ɛ4 non-carriers. Conclusions Utilizing an approach that integrates self-evaluation of executive functioning with objective neurocognitive assessment may help identify the earliest signs of impending cognitive decline, particularly in those with genetic risk for AD. Such an approach could sensitively determine those most prone to future cognitive decline prior to symptom onset, when interventions could be most effective.
Collapse
Affiliation(s)
- Sarah A. Evans
- Department of Psychology, Marquette University, Milwaukee, WI, USA
| | | | - Riya Bhasin
- Department of Psychology, Marquette University, Milwaukee, WI, USA
| | | |
Collapse
|
35
|
Bakhtiari A, Benedek K, Law I, Fagerlund B, Mortensen EL, Osler M, Lauritzen M, Larsson HBW, Vestergaard MB. Early cerebral amyloid-β accumulation and hypermetabolism are associated with subtle cognitive deficits before accelerated cerebral atrophy. GeroScience 2024; 46:769-782. [PMID: 38102439 PMCID: PMC10828321 DOI: 10.1007/s11357-023-01031-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 12/02/2023] [Indexed: 12/17/2023] Open
Abstract
AIMS Alzheimer's disease (AD) is characterized by the accumulation of amyloid beta (Aβ) in the brain. The deposition of Aβ is believed to initiate a detrimental cascade, including cerebral hypometabolism, accelerated brain atrophy, and cognitive problems-ultimately resulting in AD. However, the timing and causality of the cascade resulting in AD are not yet fully established. Therefore, we examined whether early Aβ accumulation affects cerebral glucose metabolism, atrophy rate, and age-related cognitive decline before the onset of neurodegenerative disease. METHODS Participants from the Metropolit 1953 Danish Male Birth Cohort underwent brain positron emission tomography (PET) imaging using the radiotracers [11C]Pittsburgh Compound-B (PiB) (N = 70) and [18F]Fluorodeoxyglucose (FDG) (N = 76) to assess cerebral Aβ accumulation and glucose metabolism, respectively. The atrophy rate was calculated from anatomical magnetic resonance imaging (MRI) scans conducted presently and 10 years ago. Cognitive decline was examined from neurophysiological tests conducted presently and ten or 5 years ago. RESULTS Higher Aβ accumulation in AD-critical brain regions correlated with greater visual memory decline (p = 0.023). Aβ accumulation did not correlate with brain atrophy rates. Increased cerebral glucose metabolism in AD-susceptible regions correlated with worse verbal memory performance (p = 0.040). CONCLUSIONS Aβ accumulation in known AD-related areas was associated with subtle cognitive deficits. The association was observed before hypometabolism or accelerated brain atrophy, suggesting that Aβ accumulation is involved early in age-related cognitive dysfunction. The association between hypermetabolism and worse memory performance may be due to early compensatory mechanisms adapting for malfunctioning neurons by increasing metabolism.
Collapse
Affiliation(s)
- Aftab Bakhtiari
- Functional Imaging Unit, Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet Glostrup, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.
- Department of Clinical Neurophysiology, The Neuroscience Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.
- Faculty of Health and Medical Sciences, Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark.
- Center for Healthy Aging, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Krisztina Benedek
- Department of Neurology, Neurophysiology, Zealand University Hospital, Roskilde, Denmark
| | - Ian Law
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet, , University of Copenhagen, Copenhagen, Denmark
| | - Birgitte Fagerlund
- Department of Psychology, University of Copenhagen, Copenhagen, Denmark
- Child and Adolescent Mental Health Center, Copenhagen University Hospital - Mental Health Services CPH, Copenhagen, Denmark
| | | | - Merete Osler
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Martin Lauritzen
- Department of Clinical Neurophysiology, The Neuroscience Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
- Center for Healthy Aging, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Henrik B W Larsson
- Functional Imaging Unit, Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet Glostrup, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Faculty of Health and Medical Sciences, Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Mark B Vestergaard
- Functional Imaging Unit, Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet Glostrup, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
36
|
Lopez OL, Villemagne VL, Chang YF, Cohen AD, Klunk WE, Mathis CA, Pascoal T, Ikonomovic MD, Rowe C, Dore V, Snitz BE, Lopresti BJ, Kamboh MI, Aizenstein HJ, Kuller LH. Association Between β-Amyloid Accumulation and Incident Dementia in Individuals 80 Years or Older Without Dementia. Neurology 2024; 102:e207920. [PMID: 38165336 PMCID: PMC10870745 DOI: 10.1212/wnl.0000000000207920] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 10/03/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND AND OBJECTIVES While the highest prevalence of dementia occurs in individuals older than 80 years, most imaging studies focused on younger populations. The rates of β-amyloid (Aβ) accumulation and the effect of Alzheimer disease (AD) pathology on progression to dementia in this age group remain unexplored. In this study, we examined the relationship between changes in Aβ deposition over time and incident dementia in nondemented individuals followed during a period of 11 years. METHODS We examined 94 participants (age 85.9 + 2.8 years) who had up to 5 measurements of Pittsburgh compound-B (PiB)-PET and clinical evaluations from 2009 to 2020. All 94 participants had 2 PiB-PET scans, 76 participants had 3 PiB-PET scans, 18 participants had 4 PiB-PET scans, and 10 participants had 5 PiB-PET scans. The rates of Aβ deposition were compared with 120 nondemented individuals younger than 80 years (69.3 ± 5.4 years) from the Australian Imaging, Biomarker, and Lifestyle (AIBL) study who had 3 or more annual PiB-PET assessments. RESULTS By 2020, 49% of the participants developed dementia and 63% were deceased. There was a gradual increase in Aβ deposition in all participants whether they were considered Aβ positive or negative at baseline. In a Cox model controlled for age, sex, education level, APOE-4 allele, baseline Mini-Mental State Examination, and mortality, short-term change in Aβ deposition was not significantly associated with incident dementia (HR 2.19 (0.41-11.73). However, baseline Aβ burden, cortical thickness, and white matter lesions volume were the predictors of incident dementia. Aβ accumulation was faster (p = 0.01) in the older cohort (5.6%/year) when compared with AIBL (4.1%/year). In addition, baseline Aβ deposition was a predictor of short-term change (mean time 1.88 years). DISCUSSION There was an accelerated Aβ accumulation in cognitively normal individuals older than 80 years. Baseline Aβ deposition was a determinant of incident dementia and short-term change in Aβ deposition suggesting that an active Aβ pathologic process was present when these participants were cognitively normal. Consequently, age may not be a limiting factor for the use of the emergent anti-Aβ therapies.
Collapse
Affiliation(s)
- Oscar L Lopez
- From the Departments of Neurology (O.L.L., W.E.K., M.D.I., B.E.S.), Psychiatry (O.L.L., V.L.V., A.D.C., W.E.K., T.P., H.J.A.), Neurosurgery (Y.-F.C.), Radiology (A.D.C., C.A.M., B.J.L.), Epidemiology (L.H.K.), and Human Genetics, Graduate School of Public Health (M.I.K.), University of Pittsburgh, PA; Department of Molecular Imaging and Therapy (C.R.), Austin Health, Melbourne; The Florey Institute of Neuroscience and Mental Health (C.R., V.D.), University of Melbourne; and CSIRO Health and Biosecurity (V.D.), Melbourne, Australia
| | - Victor L Villemagne
- From the Departments of Neurology (O.L.L., W.E.K., M.D.I., B.E.S.), Psychiatry (O.L.L., V.L.V., A.D.C., W.E.K., T.P., H.J.A.), Neurosurgery (Y.-F.C.), Radiology (A.D.C., C.A.M., B.J.L.), Epidemiology (L.H.K.), and Human Genetics, Graduate School of Public Health (M.I.K.), University of Pittsburgh, PA; Department of Molecular Imaging and Therapy (C.R.), Austin Health, Melbourne; The Florey Institute of Neuroscience and Mental Health (C.R., V.D.), University of Melbourne; and CSIRO Health and Biosecurity (V.D.), Melbourne, Australia
| | - Yue-Fang Chang
- From the Departments of Neurology (O.L.L., W.E.K., M.D.I., B.E.S.), Psychiatry (O.L.L., V.L.V., A.D.C., W.E.K., T.P., H.J.A.), Neurosurgery (Y.-F.C.), Radiology (A.D.C., C.A.M., B.J.L.), Epidemiology (L.H.K.), and Human Genetics, Graduate School of Public Health (M.I.K.), University of Pittsburgh, PA; Department of Molecular Imaging and Therapy (C.R.), Austin Health, Melbourne; The Florey Institute of Neuroscience and Mental Health (C.R., V.D.), University of Melbourne; and CSIRO Health and Biosecurity (V.D.), Melbourne, Australia
| | - Ann D Cohen
- From the Departments of Neurology (O.L.L., W.E.K., M.D.I., B.E.S.), Psychiatry (O.L.L., V.L.V., A.D.C., W.E.K., T.P., H.J.A.), Neurosurgery (Y.-F.C.), Radiology (A.D.C., C.A.M., B.J.L.), Epidemiology (L.H.K.), and Human Genetics, Graduate School of Public Health (M.I.K.), University of Pittsburgh, PA; Department of Molecular Imaging and Therapy (C.R.), Austin Health, Melbourne; The Florey Institute of Neuroscience and Mental Health (C.R., V.D.), University of Melbourne; and CSIRO Health and Biosecurity (V.D.), Melbourne, Australia
| | - William E Klunk
- From the Departments of Neurology (O.L.L., W.E.K., M.D.I., B.E.S.), Psychiatry (O.L.L., V.L.V., A.D.C., W.E.K., T.P., H.J.A.), Neurosurgery (Y.-F.C.), Radiology (A.D.C., C.A.M., B.J.L.), Epidemiology (L.H.K.), and Human Genetics, Graduate School of Public Health (M.I.K.), University of Pittsburgh, PA; Department of Molecular Imaging and Therapy (C.R.), Austin Health, Melbourne; The Florey Institute of Neuroscience and Mental Health (C.R., V.D.), University of Melbourne; and CSIRO Health and Biosecurity (V.D.), Melbourne, Australia
| | - Chester A Mathis
- From the Departments of Neurology (O.L.L., W.E.K., M.D.I., B.E.S.), Psychiatry (O.L.L., V.L.V., A.D.C., W.E.K., T.P., H.J.A.), Neurosurgery (Y.-F.C.), Radiology (A.D.C., C.A.M., B.J.L.), Epidemiology (L.H.K.), and Human Genetics, Graduate School of Public Health (M.I.K.), University of Pittsburgh, PA; Department of Molecular Imaging and Therapy (C.R.), Austin Health, Melbourne; The Florey Institute of Neuroscience and Mental Health (C.R., V.D.), University of Melbourne; and CSIRO Health and Biosecurity (V.D.), Melbourne, Australia
| | - Tharick Pascoal
- From the Departments of Neurology (O.L.L., W.E.K., M.D.I., B.E.S.), Psychiatry (O.L.L., V.L.V., A.D.C., W.E.K., T.P., H.J.A.), Neurosurgery (Y.-F.C.), Radiology (A.D.C., C.A.M., B.J.L.), Epidemiology (L.H.K.), and Human Genetics, Graduate School of Public Health (M.I.K.), University of Pittsburgh, PA; Department of Molecular Imaging and Therapy (C.R.), Austin Health, Melbourne; The Florey Institute of Neuroscience and Mental Health (C.R., V.D.), University of Melbourne; and CSIRO Health and Biosecurity (V.D.), Melbourne, Australia
| | - Milos D Ikonomovic
- From the Departments of Neurology (O.L.L., W.E.K., M.D.I., B.E.S.), Psychiatry (O.L.L., V.L.V., A.D.C., W.E.K., T.P., H.J.A.), Neurosurgery (Y.-F.C.), Radiology (A.D.C., C.A.M., B.J.L.), Epidemiology (L.H.K.), and Human Genetics, Graduate School of Public Health (M.I.K.), University of Pittsburgh, PA; Department of Molecular Imaging and Therapy (C.R.), Austin Health, Melbourne; The Florey Institute of Neuroscience and Mental Health (C.R., V.D.), University of Melbourne; and CSIRO Health and Biosecurity (V.D.), Melbourne, Australia
| | - Christopher Rowe
- From the Departments of Neurology (O.L.L., W.E.K., M.D.I., B.E.S.), Psychiatry (O.L.L., V.L.V., A.D.C., W.E.K., T.P., H.J.A.), Neurosurgery (Y.-F.C.), Radiology (A.D.C., C.A.M., B.J.L.), Epidemiology (L.H.K.), and Human Genetics, Graduate School of Public Health (M.I.K.), University of Pittsburgh, PA; Department of Molecular Imaging and Therapy (C.R.), Austin Health, Melbourne; The Florey Institute of Neuroscience and Mental Health (C.R., V.D.), University of Melbourne; and CSIRO Health and Biosecurity (V.D.), Melbourne, Australia
| | - Vincent Dore
- From the Departments of Neurology (O.L.L., W.E.K., M.D.I., B.E.S.), Psychiatry (O.L.L., V.L.V., A.D.C., W.E.K., T.P., H.J.A.), Neurosurgery (Y.-F.C.), Radiology (A.D.C., C.A.M., B.J.L.), Epidemiology (L.H.K.), and Human Genetics, Graduate School of Public Health (M.I.K.), University of Pittsburgh, PA; Department of Molecular Imaging and Therapy (C.R.), Austin Health, Melbourne; The Florey Institute of Neuroscience and Mental Health (C.R., V.D.), University of Melbourne; and CSIRO Health and Biosecurity (V.D.), Melbourne, Australia
| | - Beth E Snitz
- From the Departments of Neurology (O.L.L., W.E.K., M.D.I., B.E.S.), Psychiatry (O.L.L., V.L.V., A.D.C., W.E.K., T.P., H.J.A.), Neurosurgery (Y.-F.C.), Radiology (A.D.C., C.A.M., B.J.L.), Epidemiology (L.H.K.), and Human Genetics, Graduate School of Public Health (M.I.K.), University of Pittsburgh, PA; Department of Molecular Imaging and Therapy (C.R.), Austin Health, Melbourne; The Florey Institute of Neuroscience and Mental Health (C.R., V.D.), University of Melbourne; and CSIRO Health and Biosecurity (V.D.), Melbourne, Australia
| | - Brian J Lopresti
- From the Departments of Neurology (O.L.L., W.E.K., M.D.I., B.E.S.), Psychiatry (O.L.L., V.L.V., A.D.C., W.E.K., T.P., H.J.A.), Neurosurgery (Y.-F.C.), Radiology (A.D.C., C.A.M., B.J.L.), Epidemiology (L.H.K.), and Human Genetics, Graduate School of Public Health (M.I.K.), University of Pittsburgh, PA; Department of Molecular Imaging and Therapy (C.R.), Austin Health, Melbourne; The Florey Institute of Neuroscience and Mental Health (C.R., V.D.), University of Melbourne; and CSIRO Health and Biosecurity (V.D.), Melbourne, Australia
| | - M Ilyas Kamboh
- From the Departments of Neurology (O.L.L., W.E.K., M.D.I., B.E.S.), Psychiatry (O.L.L., V.L.V., A.D.C., W.E.K., T.P., H.J.A.), Neurosurgery (Y.-F.C.), Radiology (A.D.C., C.A.M., B.J.L.), Epidemiology (L.H.K.), and Human Genetics, Graduate School of Public Health (M.I.K.), University of Pittsburgh, PA; Department of Molecular Imaging and Therapy (C.R.), Austin Health, Melbourne; The Florey Institute of Neuroscience and Mental Health (C.R., V.D.), University of Melbourne; and CSIRO Health and Biosecurity (V.D.), Melbourne, Australia
| | - Howard J Aizenstein
- From the Departments of Neurology (O.L.L., W.E.K., M.D.I., B.E.S.), Psychiatry (O.L.L., V.L.V., A.D.C., W.E.K., T.P., H.J.A.), Neurosurgery (Y.-F.C.), Radiology (A.D.C., C.A.M., B.J.L.), Epidemiology (L.H.K.), and Human Genetics, Graduate School of Public Health (M.I.K.), University of Pittsburgh, PA; Department of Molecular Imaging and Therapy (C.R.), Austin Health, Melbourne; The Florey Institute of Neuroscience and Mental Health (C.R., V.D.), University of Melbourne; and CSIRO Health and Biosecurity (V.D.), Melbourne, Australia
| | - Lewis H Kuller
- From the Departments of Neurology (O.L.L., W.E.K., M.D.I., B.E.S.), Psychiatry (O.L.L., V.L.V., A.D.C., W.E.K., T.P., H.J.A.), Neurosurgery (Y.-F.C.), Radiology (A.D.C., C.A.M., B.J.L.), Epidemiology (L.H.K.), and Human Genetics, Graduate School of Public Health (M.I.K.), University of Pittsburgh, PA; Department of Molecular Imaging and Therapy (C.R.), Austin Health, Melbourne; The Florey Institute of Neuroscience and Mental Health (C.R., V.D.), University of Melbourne; and CSIRO Health and Biosecurity (V.D.), Melbourne, Australia
| |
Collapse
|
37
|
Guo ZX, Liu F, Wang FY, Ou YN, Huang LY, Hu H, Wang ZB, Fu Y, Gao PY, Tan L, Yu JT. CAIDE Score, Alzheimer's Disease Pathology, and Cognition in Cognitively Normal Adults: The CABLE Study. J Alzheimers Dis 2024; 99:1273-1283. [PMID: 38728186 DOI: 10.3233/jad-240005] [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: 05/12/2024]
Abstract
Background Cardiovascular Risk Factors, Ageing and Dementia (CAIDE) risk score serves as a credible predictor of an individual's risk of dementia. However, studies on the link of the CAIDE score to Alzheimer's disease (AD) pathology are scarce. Objective To explore the links of CAIDE score to cerebrospinal fluid (CSF) biomarkers of AD as well as to cognitive performance. Methods In the Chinese Alzheimer's Biomarker and LifestylE (CABLE) study, we recruited 600 cognitively normal participants. Correlations between the CAIDE score and CSF biomarkers of AD as well as cognitive performance were probed through multiple linear regression models. Whether the correlation between CAIDE score and cognitive performance was mediated by AD pathology was researched by means of mediation analyses. Results Linear regression analyses illustrated that CAIDE score was positively associated with tau-related biomarkers, including pTau (p < 0.001), tTau (p < 0.001), as well as tTau/Aβ42 (p = 0.008), while it was in negative association with cognitive scores, consisting of MMSE score (p < 0.001) as well as MoCA score (p < 0.001). The correlation from CAIDE score to cognitive scores was in part mediated by tau pathology, with a mediation rate varying from 3.2% to 13.2%. Conclusions A higher CAIDE score, as demonstrated in our study, was linked to more severe tau pathology and poorer cognitive performance, and tau pathology mediated the link of CAIDE score to cognitive performance. Increased dementia risk will lead to cognitive decline through aggravating neurodegeneration.
Collapse
Affiliation(s)
- Ze-Xin Guo
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Fang Liu
- Shandong Xiehe University, Jinan, Shandong, China
| | - Fang-Yuan Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Liang-Yu Huang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Hao Hu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Zhi-Bo Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yan Fu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Pei-Yang Gao
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
38
|
Loreto F, Verdi S, Kia SM, Duvnjak A, Hakeem H, Fitzgerald A, Patel N, Lilja J, Win Z, Perry R, Marquand AF, Cole JH, Malhotra P. Alzheimer's disease heterogeneity revealed by neuroanatomical normative modeling. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12559. [PMID: 38487076 PMCID: PMC10937817 DOI: 10.1002/dad2.12559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 10/11/2023] [Accepted: 01/30/2024] [Indexed: 03/17/2024]
Abstract
INTRODUCTION Overlooking the heterogeneity in Alzheimer's disease (AD) may lead to diagnostic delays and failures. Neuroanatomical normative modeling captures individual brain variation and may inform our understanding of individual differences in AD-related atrophy. METHODS We applied neuroanatomical normative modeling to magnetic resonance imaging from a real-world clinical cohort with confirmed AD (n = 86). Regional cortical thickness was compared to a healthy reference cohort (n = 33,072) and the number of outlying regions was summed (total outlier count) and mapped at individual- and group-levels. RESULTS The superior temporal sulcus contained the highest proportion of outliers (60%). Elsewhere, overlap between patient atrophy patterns was low. Mean total outlier count was higher in patients who were non-amnestic, at more advanced disease stages, and without depressive symptoms. Amyloid burden was negatively associated with outlier count. DISCUSSION Brain atrophy in AD is highly heterogeneous and neuroanatomical normative modeling can be used to explore anatomo-clinical correlations in individual patients.
Collapse
Affiliation(s)
- Flavia Loreto
- Department of Brain SciencesFaculty of MedicineImperial College LondonLondonUK
| | - Serena Verdi
- Centre for Medical Image ComputingMedical Physics and Biomedical EngineeringUniversity College LondonLondonUK
- Dementia Research CentreUCL Queen Square Institute of NeurologyLondonUK
| | - Seyed Mostafa Kia
- Donders Centre for Cognitive NeuroimagingDonders Institute for BrainCognition and BehaviourRadboud UniversityNijmegenThe Netherlands
- Department of Cognitive NeuroscienceRadboud University Medical CentreNijmegenThe Netherlands
- Department of PsychiatryUtrecht University Medical CenterUtrechtThe Netherlands
| | - Aleksandar Duvnjak
- Department of Brain SciencesFaculty of MedicineImperial College LondonLondonUK
| | - Haneen Hakeem
- Department of Brain SciencesFaculty of MedicineImperial College LondonLondonUK
| | - Anna Fitzgerald
- Department of Brain SciencesFaculty of MedicineImperial College LondonLondonUK
| | - Neva Patel
- Department of Nuclear MedicineImperial College Healthcare NHS TrustLondonUK
| | | | - Zarni Win
- Department of Nuclear MedicineImperial College Healthcare NHS TrustLondonUK
| | - Richard Perry
- Department of Brain SciencesFaculty of MedicineImperial College LondonLondonUK
- Department of NeurologyImperial College Healthcare NHS TrustLondonUK
| | - Andre F. Marquand
- Donders Centre for Cognitive NeuroimagingDonders Institute for BrainCognition and BehaviourRadboud UniversityNijmegenThe Netherlands
- Department of Cognitive NeuroscienceRadboud University Medical CentreNijmegenThe Netherlands
| | - James H. Cole
- Centre for Medical Image ComputingMedical Physics and Biomedical EngineeringUniversity College LondonLondonUK
- Dementia Research CentreUCL Queen Square Institute of NeurologyLondonUK
| | - Paresh Malhotra
- Department of Brain SciencesFaculty of MedicineImperial College LondonLondonUK
- Department of NeurologyImperial College Healthcare NHS TrustLondonUK
- UK Dementia Research Institute Care Research and Technology CentreImperial College London and the University of SurreyLondonUK
| |
Collapse
|
39
|
Pietilä E, Snellman A, Tuisku J, Helin S, Viitanen M, Jula A, Rinne JO, Ekblad LL. Midlife insulin resistance, APOE genotype, and change in late-life brain beta-amyloid accumulation - A 5-year follow-up [ 11C]PIB-PET study. Neurobiol Dis 2024; 190:106385. [PMID: 38123104 DOI: 10.1016/j.nbd.2023.106385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/14/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
We studied if midlife insulin resistance (IR) and APOE genotype would predict brain beta-amyloid (Aβ) accumulation and Aβ change in late-life in 5-year follow-up [11C]PIB-PET study. 43 dementia-free participants were scanned twice with [11C]PIB-PET in their late-life (mean age at follow-up 75.4 years). Participants were recruited from the Finnish Health2000 study according to their HOMA-IR values measured in midlife (mean age at midlife 55.4 years; IR+ group, HOMA-IR > 2.17; IR- group, HOMA-IR <1.25), and their APOEε4 genotype. At late-life follow-up, [11C]PIB-PET composite SUVr was significantly higher in IR+ group than IR- group (median 2.3 (interquartile range 1.7-3.3) vs. 1.7 (1.5-2.4), p = 0.03). There was no difference between IR- and IR+ groups in [11C]PIB-PET SUVr 5-year change, but the change was significantly higher in IR+/APOEε4+ group (median change 0.8 (0.60-1.0)) than in IR-/APOEε4- (0.28 (0.14-0.47), p = 0.02) and in IR+/APOEε4- group (0.24 (0.06-0.40), p = 0.046). These results suggest that APOEε4 carriers with midlife IR are at increased risk for late-life Aβ accumulation.
Collapse
Affiliation(s)
- Elina Pietilä
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland.
| | - Anniina Snellman
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Jouni Tuisku
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Semi Helin
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Matti Viitanen
- Department of Geriatrics, Turku City Hospital and University of Turku, Finland; Division of Clinical Geriatrics, NVS, Karolinska Institutet, Stockholm, Sweden
| | - Antti Jula
- Finnish Institute for Health and Welfare, Turku, Finland
| | - Juha O Rinne
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland; InFLAMES Reseach Flagship Center, University of Turku, Turku, Finland
| | - Laura L Ekblad
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland; Department of Geriatrics, Turku University Hospital, Wellbeing services county of Southwestern Finland, Finland
| |
Collapse
|
40
|
Gao PY, Ou YN, Huang YM, Wang ZB, Fu Y, Ma YH, Li QY, Ma LY, Cui RP, Mi YC, Tan L, Yu JT. Associations between liver function and cerebrospinal fluid biomarkers of Alzheimer's disease pathology in non-demented adults: The CABLE study. J Neurochem 2024; 168:39-51. [PMID: 38055867 DOI: 10.1111/jnc.16025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 11/16/2023] [Accepted: 11/21/2023] [Indexed: 12/08/2023]
Abstract
Liver function has been suggested as a possible factor in the progression of Alzheimer's disease (AD) development. However, the association between liver function and cerebrospinal fluid (CSF) levels of AD biomarkers remains unclear. In this study, we analyzed the data from 1687 adults without dementia from the Chinese Alzheimer's Biomarker and LifestylE study to investigate differences in liver function between pathological and clinical AD groups, as defined by the 2018 National Institute on Aging-Alzheimer's Association Research Framework. We also examined the linear relationship between liver function, CSF AD biomarkers, and cognition using linear regression models. Furthermore, mediation analyses were applied to explore the potential mediation effects of AD pathological biomarkers on cognition. Our findings indicated that, with AD pathological and clinical progression, the concentrations of total protein (TP), globulin (GLO), and aspartate aminotransferase/alanine transaminase (ALT) increased, while albumin/globulin (A/G), adenosine deaminase, alpha-L-fucosidase, albumin, prealbumin, ALT, and glutamate dehydrogenase (GLDH) concentrations decreased. Furthermore, we also identified significant relationships between TP (β = -0.115, pFDR < 0.001), GLO (β = -0.184, pFDR < 0.001), and A/G (β = 0.182, pFDR < 0.001) and CSF β-amyloid1-42 (Aβ1-42 ) (and its related CSF AD biomarkers). Moreover, after 10 000 bootstrapped iterations, we identified a potential mechanism by which TP and GLDH may affect cognition by mediating CSF AD biomarkers, with mediation effect sizes ranging from 3.91% to 16.44%. Overall, our results suggested that abnormal liver function might be involved in the clinical and pathological progression of AD. Amyloid and tau pathologies also might partially mediate the relationship between liver function and cognition. Future research is needed to fully understand the underlying mechanisms and causality to develop an approach to AD prevention and treatment approach.
Collapse
Affiliation(s)
- Pei-Yang Gao
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yi-Ming Huang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Zhi-Bo Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, China
| | - Yan Fu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Hui Ma
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Qiong-Yao Li
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Li-Yun Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Rui-Ping Cui
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yin-Chu Mi
- Department of Neurology, Qingdao Municipal Hospital, Dalian Medical University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
41
|
Rani N, Alm KH, Corona-Long CA, Speck CL, Soldan A, Pettigrew C, Zhu Y, Albert M, Bakker A. Tau PET burden in Brodmann areas 35 and 36 is associated with individual differences in cognition in non-demented older adults. Front Aging Neurosci 2023; 15:1272946. [PMID: 38161595 PMCID: PMC10757623 DOI: 10.3389/fnagi.2023.1272946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/23/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction The accumulation of neurofibrillary tau tangles, a neuropathological hallmark of Alzheimer's disease (AD), occurs in medial temporal lobe (MTL) regions early in the disease process, with some of the earliest deposits localized to subregions of the entorhinal cortex. Although functional specialization of entorhinal cortex subregions has been reported, few studies have considered functional associations with localized tau accumulation. Methods In this study, stepwise linear regressions were used to examine the contributions of regional tau burden in specific MTL subregions, as measured by 18F-MK6240 PET, to individual variability in cognition. Dependent measures of interest included the Clinical Dementia Rating Sum of Boxes (CDR-SB), Mini Mental State Examination (MMSE), and composite scores of delayed episodic memory and language. Other model variables included age, sex, education, APOE4 status, and global amyloid burden, indexed by 11C-PiB. Results Tau burden in right Brodmann area 35 (BA35), left and right Brodmann area 36 (BA36), and age each uniquely contributed to the proportion of explained variance in CDR-SB scores, while right BA36 and age were also significant predictors of MMSE scores, and right BA36 was significantly associated with delayed episodic memory performance. Tau burden in both left and right BA36, along with education, uniquely contributed to the proportion of explained variance in language composite scores. Importantly, the addition of more inclusive ROIs, encompassing less granular segmentation of the entorhinal cortex, did not significantly contribute to explained variance in cognition across any of the models. Discussion These findings suggest that the ability to quantify tau burden in more refined MTL subregions may better account for individual differences in cognition, which may improve the identification of non-demented older adults who are on a trajectory of decline due to AD.
Collapse
Affiliation(s)
- Nisha Rani
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kylie H. Alm
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Caitlin A. Corona-Long
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Caroline L. Speck
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Anja Soldan
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Corinne Pettigrew
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yuxin Zhu
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Marilyn Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Arnold Bakker
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
42
|
Xia Y, Maruff P, Doré V, Bourgeat P, Laws SM, Fowler C, Rainey-Smith SR, Martins RN, Villemagne VL, Rowe CC, Masters CL, Coulson EJ, Fripp J. Longitudinal trajectories of basal forebrain volume in normal aging and Alzheimer's disease. Neurobiol Aging 2023; 132:120-130. [PMID: 37801885 DOI: 10.1016/j.neurobiolaging.2023.09.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 08/03/2023] [Accepted: 09/07/2023] [Indexed: 10/08/2023]
Abstract
Dysfunction of the cholinergic basal forebrain (BF) system and amyloid-β (Aβ) deposition are early pathological features in Alzheimer's disease (AD). However, their association in early AD is not well-established. This study investigated the nature and magnitude of volume loss in the BF, over an extended period, in 516 older adults who completed Aβ-PET and serial magnetic resonance imaging scans. Individuals were grouped at baseline according to the presence of cognitive impairment (CU, CI) and Aβ status (Aβ-, Aβ+). Longitudinal volumetric changes in the BF and hippocampus were assessed across groups. The results indicated that high Aβ levels correlated with faster volume loss in the BF and hippocampus, and the effect of Aβ varied within BF subregions. Compared to CU Aβ+ individuals, Aβ-related loss among CI Aβ+ adults was much greater in the predominantly cholinergic subregion of Ch4p, whereas no difference was observed for the Ch1/Ch2 region. The findings support early and substantial vulnerability of the BF and further reveal distinctive degeneration of BF subregions during early AD.
Collapse
Affiliation(s)
- Ying Xia
- The Australian e-Health Research Centre, CSIRO Health and Biosecurity, Brisbane, Queensland, Australia.
| | - Paul Maruff
- Cogstate Ltd, Melbourne, Victoria, Australia; The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Vincent Doré
- Department of Nuclear Medicine and Centre for PET, Austin Health, Melbourne, Victoria, Australia; The Australian e-Health Research Centre, CSIRO Health and Biosecurity, Melbourne, Victoria, Australia
| | - Pierrick Bourgeat
- The Australian e-Health Research Centre, CSIRO Health and Biosecurity, Brisbane, Queensland, Australia
| | - Simon M Laws
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia; Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia; Curtin Medical School, Curtin University, Bentley, Western Australia, Australia
| | - Christopher Fowler
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Stephanie R Rainey-Smith
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, Western Australia, Australia; Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia; School of Psychological Science, University of Western Australia, Crawley, Western Australia, Australia; School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Ralph N Martins
- Australian Alzheimer's Research Foundation, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia; School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia; Department of Biomedical Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Victor L Villemagne
- Department of Nuclear Medicine and Centre for PET, Austin Health, Melbourne, Victoria, Australia; Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Christopher C Rowe
- Department of Nuclear Medicine and Centre for PET, Austin Health, Melbourne, Victoria, Australia; Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Elizabeth J Coulson
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia; School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Jurgen Fripp
- The Australian e-Health Research Centre, CSIRO Health and Biosecurity, Brisbane, Queensland, Australia
| |
Collapse
|
43
|
Wang D, Xu K, Dang M, Sang F, Chen K, Zhang Z, Li X. Multi-domain cognition dysfunction accompanies frontoparietal and temporal amyloid accumulation in the elderly. Cereb Cortex 2023; 33:11329-11338. [PMID: 37859548 PMCID: PMC11486686 DOI: 10.1093/cercor/bhad369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/21/2023] Open
Abstract
It is helpful to understand the pathology of Alzheimer's disease by exploring the relationship between amyloid-β accumulation and cognition. The study explored the relationship between regional amyloid-β accumulation and multiple cognitions and study their application value in the Alzheimer's disease diagnosis. 135 participants completed 18F-florbetapir Positron Emission Tomography (PET), structural MRI, and a cognitive battery. Partial correlation was used to examine the relationship between global and regional amyloid-β accumulation and cognitions. Then, a support vector machine was applied to determine whether cognition-related accumulation regions can adequately distinguish the cognitively normal controls (76 participants) and mild cognitive impairment (30 participants) groups or mild cognitive impairment and Alzheimer's disease (29 participants) groups. The result showed that amyloid-β accumulation regions were mainly located in the frontoparietal cortex, calcarine fissure, and surrounding cortex and temporal pole regions. Episodic memory-related regions included the frontoparietal cortices; executive function-related regions included the frontoparietal, temporal, and occipital cortices; and processing speed-related regions included the frontal and occipital cortices. Support vector machine analysis showed that only episodic memory-related amyloid-β accumulation regions had better classification performance during the progression of Alzheimer's disease. Assessing regional changes in amyloid, particularly in frontoparietal regions, can aid in the early detection of amyloid-related decline in cognitive function.
Collapse
Affiliation(s)
- Dandan Wang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, No. 19, Xinjiekouwai St, Haidian District, Beijing, 100875, P.R. China
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Center, Beijing Normal University, No. 19, Xinjiekouwai St, Haidian District, Beijing, 100875, P.R. China
| | - Kai Xu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, No. 19, Xinjiekouwai St, Haidian District, Beijing, 100875, P.R. China
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Center, Beijing Normal University, No. 19, Xinjiekouwai St, Haidian District, Beijing, 100875, P.R. China
- School of Artificial Intelligence, Beijing Normal University, No. 19, Xinjiekouwai St, Haidian District, Beijing, 100875, P.R. China
| | - Mingxi Dang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, No. 19, Xinjiekouwai St, Haidian District, Beijing, 100875, P.R. China
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Center, Beijing Normal University, No. 19, Xinjiekouwai St, Haidian District, Beijing, 100875, P.R. China
| | - Feng Sang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, No. 19, Xinjiekouwai St, Haidian District, Beijing, 100875, P.R. China
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Center, Beijing Normal University, No. 19, Xinjiekouwai St, Haidian District, Beijing, 100875, P.R. China
| | - Kewei Chen
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Center, Beijing Normal University, No. 19, Xinjiekouwai St, Haidian District, Beijing, 100875, P.R. China
- Banner Alzheimer’s Institute, Phoenix, AZ 85006, United States
| | - Zhanjun Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, No. 19, Xinjiekouwai St, Haidian District, Beijing, 100875, P.R. China
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Center, Beijing Normal University, No. 19, Xinjiekouwai St, Haidian District, Beijing, 100875, P.R. China
| | - Xin Li
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, No. 19, Xinjiekouwai St, Haidian District, Beijing, 100875, P.R. China
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Center, Beijing Normal University, No. 19, Xinjiekouwai St, Haidian District, Beijing, 100875, P.R. China
| |
Collapse
|
44
|
Eraslan Boz H, Koçoğlu K, Akkoyun M, Tüfekci IY, Ekin M, Akdal G. Eye movement patterns during viewing face images with neutral expressions in patients with early-stage Alzheimer's disease and amnestic mild cognitive impairment. Brain Behav 2023; 13:e3232. [PMID: 37605291 PMCID: PMC10636417 DOI: 10.1002/brb3.3232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/08/2023] [Accepted: 08/10/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) neuropathology affects the brain regions responsible for visuospatial skills. Accumulating evidence points to visual difficulties involving face processing in AD and amnestic mild cognitive impairment (aMCI). No study has so far examined eye movement patterns when viewing faces with neutral expressions in patients with AD. AIM The objective of this study aimed to examine the eye movements of patients with early-stage AD, aMCI, and healthy controls (HC) during viewing face images. MATERIALS&METHODS Thirty-one AD, 37 aMCI, and 33 HC were included in the study. Eye movements in facial stimuli were recorded with the EyeLink 1000 Plus eye-tracker. RESULTS Our findings showed that AD patients looked less at the eye area of interest than the nose and mouth areas of interest compared to aMCI and HC. Regardless of the group, all participants looked at the eye and nose areas of interest more and longer in the mouth area of interest. In addition, the first fixation duration to the eye area of interest of all participants was shorter than that of the nose and mouth. DISCUSSION Consistent with our study, studies in healthy adults revealed eye movement patterns that focused more on the eyes and nose. AD patients are unable to pay attention to the salient parts of faces, tending to focus instead on the non-informative parts. CONCLUSION Our study is the first to reveal eye movement differences in face processing in AD.
Collapse
Affiliation(s)
- Hatice Eraslan Boz
- Department of Neurosciences, Institute of Health SciencesDokuz Eylül UniversityIzmirTürkiye
- Department of Neurology, Unit of NeuropsychologyDokuz Eylül UniversityIzmirTürkiye
| | - Koray Koçoğlu
- Department of Neurosciences, Institute of Health SciencesDokuz Eylül UniversityIzmirTürkiye
| | - Müge Akkoyun
- Department of Neurosciences, Institute of Health SciencesDokuz Eylül UniversityIzmirTürkiye
| | - Işıl Yağmur Tüfekci
- Department of Neurosciences, Institute of Health SciencesDokuz Eylül UniversityIzmirTürkiye
| | - Merve Ekin
- Department of Neurosciences, Institute of Health SciencesDokuz Eylül UniversityIzmirTürkiye
| | - Gülden Akdal
- Department of Neurosciences, Institute of Health SciencesDokuz Eylül UniversityIzmirTürkiye
- Department of NeurologyDokuz Eylül UniversityIzmirTürkiye
| |
Collapse
|
45
|
Sperling RA, Donohue MC, Raman R, Rafii MS, Johnson K, Masters CL, van Dyck CH, Iwatsubo T, Marshall GA, Yaari R, Mancini M, Holdridge KC, Case M, Sims JR, Aisen PS. Trial of Solanezumab in Preclinical Alzheimer's Disease. N Engl J Med 2023; 389:1096-1107. [PMID: 37458272 PMCID: PMC10559996 DOI: 10.1056/nejmoa2305032] [Citation(s) in RCA: 163] [Impact Index Per Article: 81.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
BACKGROUND Trials of monoclonal antibodies that target various forms of amyloid at different stages of Alzheimer's disease have had mixed results. METHODS We tested solanezumab, which targets monomeric amyloid, in a phase 3 trial involving persons with preclinical Alzheimer's disease. Persons 65 to 85 years of age with a global Clinical Dementia Rating score of 0 (range, 0 to 3, with 0 indicating no cognitive impairment and 3 severe dementia), a score on the Mini-Mental State Examination of 25 or more (range, 0 to 30, with lower scores indicating poorer cognition), and elevated brain amyloid levels on 18F-florbetapir positron-emission tomography (PET) were enrolled. Participants were randomly assigned in a 1:1 ratio to receive solanezumab at a dose of up to 1600 mg intravenously every 4 weeks or placebo. The primary end point was the change in the Preclinical Alzheimer Cognitive Composite (PACC) score (calculated as the sum of four z scores, with higher scores indicating better cognitive performance) over a period of 240 weeks. RESULTS A total of 1169 persons underwent randomization: 578 were assigned to the solanezumab group and 591 to the placebo group. The mean age of the participants was 72 years, approximately 60% were women, and 75% had a family history of dementia. At 240 weeks, the mean change in PACC score was -1.43 in the solanezumab group and -1.13 in the placebo group (difference, -0.30; 95% confidence interval, -0.82 to 0.22; P = 0.26). Amyloid levels on brain PET increased by a mean of 11.6 centiloids in the solanezumab group and 19.3 centiloids in the placebo group. Amyloid-related imaging abnormalities (ARIA) with edema occurred in less than 1% of the participants in each group. ARIA with microhemorrhage or hemosiderosis occurred in 29.2% of the participants in the solanezumab group and 32.8% of those in the placebo group. CONCLUSIONS Solanezumab, which targets monomeric amyloid in persons with elevated brain amyloid levels, did not slow cognitive decline as compared with placebo over a period of 240 weeks in persons with preclinical Alzheimer's disease. (Funded by the National Institute on Aging and others; A4 ClinicalTrials.gov number, NCT02008357.).
Collapse
Affiliation(s)
- Reisa A Sperling
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - Michael C Donohue
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - Rema Raman
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - Michael S Rafii
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - Keith Johnson
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - Colin L Masters
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - Christopher H van Dyck
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - Takeshi Iwatsubo
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - Gad A Marshall
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - Roy Yaari
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - Michele Mancini
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - Karen C Holdridge
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - Michael Case
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - John R Sims
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| | - Paul S Aisen
- From the Center for Alzheimer Research and Treatment, Brigham and Women's Hospital, Massachusetts General Hospital, Harvard Medical School (R.A.S., G.A.M.), and the Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School (K.J.) - both in Boston; Alzheimer's Therapeutic Research Institute, Keck School of Medicine, University of Southern California, San Diego (M.C.D., R.R., M.S.R., P.S.A.); the Florey Institute, University of Melbourne, Melbourne, VIC, Australia (C.L.M.); the Departments of Psychiatry, Neurology, and Neuroscience, Yale School of Medicine, New Haven, CT (C.H.D.); the Department of Neuropathology, Graduate School of Medicine, University of Tokyo, Tokyo (T.I.); and Eli Lilly, Indianapolis (R.Y., M.M., K.C.H., M.C., J.R.S.)
| |
Collapse
|
46
|
Atef MM, Mostafa YM, Ahmed AAM, El-Sayed NM. Simvastatin attenuates aluminium chloride-induced neurobehavioral impairments through activation of TGF-β1/ SMAD2 and GSK3β/β-catenin signalling pathways. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 102:104220. [PMID: 37454825 DOI: 10.1016/j.etap.2023.104220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/19/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterised by the presence of β-amyloid plaques and acetylcholine depletion leading to neurobehavioral defects. AD was contributed also with downregulation of TGF-β1/SMAD2 and GSK3β/β-catenin pathways. Simvastatin (SMV) improved memory function experimentally and clinically. Hence, this study aimed to investigate the mechanistic role of SMV against aluminium chloride (AlCl3) induced neurobehavioral impairments. AD was induced by AlCl3 (50 mg/kg) for 6 weeks. Mice received Simvastatin (10 or 20 mg/kg) or Donepezil (3 mg/kg) for 6 weeks after that the histopathological, immunohistochemical and biochemical test were examined. Treatment with SMV improved the memory deterioration induced by AlCl3 with significant recovery of the histopathological changes. This was concomitant with the decrease of AChE and Aβ (1-42). SMV provides its neuroprotective effect through upregulating the protein expression of β-catenin, TGF-β1 and downregulating the expression of GSK3β, TLR4 and p-SMAD2.
Collapse
Affiliation(s)
| | - Yasser M Mostafa
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt; Department of Pharmacology & Toxicology, Faculty of Pharmacy, Badr University in Cairo, Egypt
| | - Amal A M Ahmed
- Department of Cytology & Histology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Norhan M El-Sayed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt.
| |
Collapse
|
47
|
Ekblad LL, Tuisku J, Koivumäki M, Helin S, Rinne JO, Snellman A. Insulin resistance and body mass index are associated with TSPO PET in cognitively unimpaired elderly. J Cereb Blood Flow Metab 2023; 43:1588-1600. [PMID: 37113066 PMCID: PMC10414007 DOI: 10.1177/0271678x231172519] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/27/2023] [Accepted: 04/01/2023] [Indexed: 04/29/2023]
Abstract
Metabolic risk factors are associated with peripheral low-grade inflammation and an increased risk for dementia. We evaluated if metabolic risk factors i.e. insulin resistance, body mass index (BMI), serum cholesterol values, or high sensitivity C-reactive protein associate with central inflammation or beta-amyloid (Aβ) accumulation in the brain, and if these associations are modulated by APOE4 gene dose. Altogether 60 cognitively unimpaired individuals (mean age 67.7 years (SD 4.7); 63% women; 21 APOE3/3, 20 APOE3/4 and 19 APOE4/4) underwent positron emission tomography with [11C]PK11195 targeting TSPO (18 kDa translocator protein) and [11C]PIB targeting fibrillar Aβ. [11C]PK11195 distribution value ratios and [11C]PIB standardized uptake values were calculated in a cortical composite region of interest typical for Aβ accumulation in Alzheimer's disease. Associations between metabolic risk factors, [11C]PK11195, and [11C]PIB uptake were evaluated with linear models adjusted for age and sex. Higher logarithmic HOMA-IR (standardized beta 0.40, p = 0.002) and BMI (standardized beta 0.27, p = 0.048) were associated with higher TSPO availability. Voxel-wise analyses indicated that this association was mainly seen in the parietal cortex. Higher logarithmic HOMA-IR was associated with higher [11C]PIB (standardized beta 0.44, p = 0.02), but only in APOE4/4 homozygotes. BMI and HOMA-IR seem to influence TSPO availability in the brain.
Collapse
Affiliation(s)
- Laura L Ekblad
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Jouni Tuisku
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Mikko Koivumäki
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Semi Helin
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Juha O Rinne
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
- InFLAMES Reseach Flagship Center, University of Turku, Turku, Finland
| | - Anniina Snellman
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| |
Collapse
|
48
|
Xu C, Zhao L, Dong C. The performance of plasma phosphorylated tau231 in detecting Alzheimer's disease: A systematic review with meta-analysis. Eur J Neurosci 2023; 58:3132-3149. [PMID: 37501373 DOI: 10.1111/ejn.16085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/15/2023] [Accepted: 06/26/2023] [Indexed: 07/29/2023]
Abstract
Cerebrospinal fluid (CSF) phosphorylated tau231 (P-tau231) is associated with neuropathological outcomes of Alzheimer's disease (AD). The invasive access of cerebrospinal fluid has greatly stimulated interest in the identification of blood-based P-tau231, and the recent advent of single-molecule array assay for the quantification of plasma P-tau231 may provide a turning point to evaluate the usefulness of P-tau231 as an AD-related biomarker. Yet, in the plasma P-tau231 literature, findings with regard to its diagnostic utility have been inconsistent, and thus, we aimed to statistically investigate the potential of plasma P-tau231 in the context of AD via meta-analysis. Publications on plasma P-tau231 were systematically retrieved from PubMed, EMBASE, the Cochrane library and Web of Science databases. A total of 10 studies covering 2007 participants were included, and we conducted random-effect or fixed-effect meta-analysis, sensitivity analysis and publication bias analysis using the STATA SE 14.0 software. According to our quantitative integration, plasma P-tau231 increased from cognitively unimpaired (CU) populations to mild cognitive impairment to AD and showed significant changes in pairwise comparisons of AD, mild cognitive impairment and CU. Plasma P-tau231 level was significantly higher in CU controls with positive amyloid-β (Aβ) status compared with Aβ-negative CU group. Additionally, the excellent diagnostic accuracy of plasma P-tau231 for asymptomatic Aβ pathology was verified by the pooled value of area under the receiver operating characteristic curves (standard mean difference [95% confidence interval]: .75 [.69, .81], P < 0.00001). Overall, the increased plasma P-tau231 concentrations were found in relation to the early development and progression of AD.
Collapse
Affiliation(s)
- Chang Xu
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Li Zhao
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Chunbo Dong
- Department of Neurology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
49
|
Azevedo T, Bethlehem RAI, Whiteside DJ, Swaddiwudhipong N, Rowe JB, Lió P, Rittman T. Identifying healthy individuals with Alzheimer's disease neuroimaging phenotypes in the UK Biobank. COMMUNICATIONS MEDICINE 2023; 3:100. [PMID: 37474615 PMCID: PMC10359360 DOI: 10.1038/s43856-023-00313-w] [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: 01/07/2022] [Accepted: 06/05/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND Identifying prediagnostic neurodegenerative disease is a critical issue in neurodegenerative disease research, and Alzheimer's disease (AD) in particular, to identify populations suitable for preventive and early disease-modifying trials. Evidence from genetic and other studies suggests the neurodegeneration of Alzheimer's disease measured by brain atrophy starts many years before diagnosis, but it is unclear whether these changes can be used to reliably detect prediagnostic sporadic disease. METHODS We trained a Bayesian machine learning neural network model to generate a neuroimaging phenotype and AD score representing the probability of AD using structural MRI data in the Alzheimer's Disease Neuroimaging Initiative (ADNI) Cohort (cut-off 0.5, AUC 0.92, PPV 0.90, NPV 0.93). We go on to validate the model in an independent real-world dataset of the National Alzheimer's Coordinating Centre (AUC 0.74, PPV 0.65, NPV 0.80) and demonstrate the correlation of the AD-score with cognitive scores in those with an AD-score above 0.5. We then apply the model to a healthy population in the UK Biobank study to identify a cohort at risk for Alzheimer's disease. RESULTS We show that the cohort with a neuroimaging Alzheimer's phenotype has a cognitive profile in keeping with Alzheimer's disease, with strong evidence for poorer fluid intelligence, and some evidence of poorer numeric memory, reaction time, working memory, and prospective memory. We found some evidence in the AD-score positive cohort for modifiable risk factors of hypertension and smoking. CONCLUSIONS This approach demonstrates the feasibility of using AI methods to identify a potentially prediagnostic population at high risk for developing sporadic Alzheimer's disease.
Collapse
Affiliation(s)
- Tiago Azevedo
- Department of Computer Science and Technology, University of Cambridge, Cambridge, UK
| | - Richard A I Bethlehem
- Brain Mapping Unit, Department of Psychiatry, University of Cambridge, Cambridge, UK
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - David J Whiteside
- Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, UK
| | - Nol Swaddiwudhipong
- Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, UK
| | - James B Rowe
- Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, UK
| | - Pietro Lió
- Department of Computer Science and Technology, University of Cambridge, Cambridge, UK
| | - Timothy Rittman
- Department of Clinical Neurosciences and Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, UK.
| |
Collapse
|
50
|
Alchera N, Garibotto V, Tomczyk S, Treyer V, Hock C, Gietl AF, Lövblad KO, Scheffler M, Chincarini A, Frisoni GB, Ribaldi F. Patterns of amyloid accumulation in amyloid-negative cases. Neurobiol Aging 2023; 129:99-108. [PMID: 37279618 DOI: 10.1016/j.neurobiolaging.2023.05.006] [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: 10/25/2022] [Revised: 05/03/2023] [Accepted: 05/03/2023] [Indexed: 06/08/2023]
Abstract
Amyloid staging models showed that regional abnormality occurs before global positivity. Several studies assumed that the trajectory of amyloid spread is homogeneous, but clinical evidence suggests that it is highly heterogeneous. We tested whether different amyloid-β (Aβ) patterns exist by applying clustering on negative scans and investigating their demographics, clinical, cognitive, and biomarkers correlates, and cognitive trajectories. 151 individuals from Geneva and Zurich cohorts with T1-MRI, negative Aβ positron emission tomography (PET,centiloid<12) and clinical assessment were included. N=123 underwent tau PET, and N=65 follow-up neuropsychological assessment. We performed k-means clustering using 33 Aβ regional Standardized Uptake Vales ratio. Demographics, clinical, cognitive, and biomarkers differences were investigated. Longitudinal cognitive changes by baseline cluster status were estimated using a linear mixed model. The cluster analysis identified two clusters: temporal predominant (TP) and cingulate predominant (CP). TP tau deposition was higher than CP. A trend for a higher cognitive decline in TP compared to CP was observed. This study suggests the existence of two Aβ deposition patterns in the earliest phases of Aβ accumulation, differently prone to tau pathology and cognitive decline.
Collapse
Affiliation(s)
- Nicola Alchera
- Istituto Nazionale di Fisica Nucleare (INFN), Genova, University of Genoa, Genoa, Italy
| | - Valentina Garibotto
- Division of Nuclear Medicine and Molecular Imaging, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland; Laboratory of Neuroimaging and Innovative Molecular Tracers (NIMTlab), Geneva University Neurocenter and Faculty of Medicine, University of Geneva, Geneva, Switzerland; Center for Biomedical imaging (CIBM), Geneva, Switzerland
| | - Szymon Tomczyk
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland
| | - Valerie Treyer
- Department of Nuclear Medicine, University Hospital of Zurich, University of Zurich, Zurich, Switzerland; Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Christoph Hock
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland; Neurimmune, Schlieren, Switzerland
| | - Anton F Gietl
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland; Department for Geriatric Psychiatry, University Hospital for Psychiatry Zurich, Zurich, Switzerland
| | - Karl-Olof Lövblad
- Division of Radiology, Geneva University Hospitals, Geneva, Switzerland
| | - Max Scheffler
- Division of Radiology, Geneva University Hospitals, Geneva, Switzerland
| | | | - Giovanni B Frisoni
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland; Geneva Memory Center, Department of Rehabilitation and Geriatrics, Geneva University Hospitals, Geneva, Switzerland
| | - Federica Ribaldi
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland; Geneva Memory Center, Department of Rehabilitation and Geriatrics, Geneva University Hospitals, Geneva, Switzerland.
| |
Collapse
|