1
|
Byeon G, Byun MS, Yi D, Ahn H, Jung G, Sohn BK, Jung JH, Chang YY, Kim K, Choi H, Kim YH, Kim YK, Kang KM, Sohn CH, Lee DY. The Effect of Cholinesterase Inhibitors on Neurodegeneration in Individuals with Amnestic Mild Cognitive Impairment. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE : THE OFFICIAL SCIENTIFIC JOURNAL OF THE KOREAN COLLEGE OF NEUROPSYCHOPHARMACOLOGY 2025; 23:256-265. [PMID: 40223260 PMCID: PMC12000676 DOI: 10.9758/cpn.24.1238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/27/2024] [Accepted: 12/31/2024] [Indexed: 04/15/2025]
Abstract
Objective Cholinesterase inhibitors (ChEIs) are effective in treating mild to moderate Alzheimer's disease (AD) dementia by compensating for acetylcholine deficiency. While their use in mild cognitive impairment (MCI) lacks strong trial support, some studies suggest they may delay neurodegeneration. This study aims to investigate ChEIs' neuroprotective effects in individuals with amnestic MCI (aMCI) using multi-modal neuroimaging, and to determine if amyloid-beta (Aβ) deposition influences these effects. Methods Longitudinal data from a cohort study were retrospectively analyzed. A total of 118 aMCI patients (ages 55- 90), who underwent baseline evaluations encompassing the assessment of ChEI use and [11C] Pittsburgh compound B-positron emission tomography (PET), were included in the analyses. All participants also received baseline and 2-year follow-up magnetic resonance imaging and [18F] fluorodeoxyglucose-PET imaging. Results The ChEI use group exhibited a significantly lesser decline in AD-signature region cerebral metabolism (AD-CM) over a 2-year period than the ChEI non-use group (B = 0.089, 95% CI: 0.030-0.149). However, there was no significant difference in the 2-year change of AD-signature region cortical thickness (AD-CT) (B = 0.032, 95% CI: -0.075 to 0.138) and hippocampal volume (B = -88.013, 95% CI: -323.900 to 147.874) between the ChEI use and non-use groups. The presence of Aβ pathology did not moderate the effect of ChEI use on AD-CM, AD-CT, or hippocampal volume. Conclusion The findings suggest that ChEIs may delay functional neurodegeneration in aMCI individuals, irrespective of the presence of amyloid pathology.
Collapse
Affiliation(s)
- Gihwan Byeon
- Department of Psychiatry, The Catholic University of Korea, Seoul St. Mary’s Hospital, Seoul, Korea
| | - Min Soo Byun
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Korea
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Seoul National University Medical Research Center, Seoul, Korea
| | - Hyejin Ahn
- Institute of Human Behavioral Medicine, Seoul National University Medical Research Center, Seoul, Korea
- Interdisciplinary Program of Cognitive Science, College of Humanities, Seoul National University, Seoul, Korea
| | - Gijung Jung
- Institute of Human Behavioral Medicine, Seoul National University Medical Research Center, Seoul, Korea
| | - Bo Kyung Sohn
- Department of Psychiatry, Sanggye Paik Hospital, College of Medicine, Inje University, Seoul, Korea
| | - Joon Hyung Jung
- Department of Neuropsychiatry, Chungbuk National University Hospital, Cheongju, Korea
| | - Yoon Young Chang
- Department of Psychiatry, Sanggye Paik Hospital, College of Medicine, Inje University, Seoul, Korea
| | - Kyungtae Kim
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Korea
| | - Hyeji Choi
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Korea
| | - Yoon Hee Kim
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, Korea
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, Seoul, Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, Seoul, Korea
| | - Dong Young Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Korea
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea
- Institute of Human Behavioral Medicine, Seoul National University Medical Research Center, Seoul, Korea
- Interdisciplinary Program of Cognitive Science, College of Humanities, Seoul National University, Seoul, Korea
| | | |
Collapse
|
2
|
Salardini E, O'Dell RS, Tchorz E, Nabulsi NB, Huang Y, Carson RE, van Dyck CH, Mecca AP. Assessment of the relationship between synaptic density and metabotropic glutamate receptors in early Alzheimer's disease: a multi-tracer PET study. Alzheimers Res Ther 2025; 17:98. [PMID: 40329311 PMCID: PMC12054321 DOI: 10.1186/s13195-025-01739-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 04/10/2025] [Indexed: 05/08/2025]
Abstract
BACKGROUND The pathological effects of amyloid β oligomers (Aβo) may be mediated through the metabotropic glutamate receptor subtype 5 (mGluR5), leading to synaptic loss in Alzheimer's disease (AD). Positron emission tomography (PET) studies of mGluR5 using [18F]FPEB indicate a reduction of receptor binding that is focused in the medial temporal lobe in AD. Synaptic loss due to AD measured through synaptic vesicle glycoprotein 2A (SV2A) quantification with [11C]UCB-J PET is also focused in the medial temporal lobe, but with clear widespread reductions is commonly AD-affected neocortical regions. In this study, we used [18F]FPEB and [11C]UCB-J PET to investigate the relationship between mGluR5 and synaptic density in early AD. METHODS Fifteen amyloid positive participants with early AD and 12 amyloid negative, cognitively normal (CN) participants underwent PET scans with both [18F]FPEB to measure mGluR5 and [11C]UCB-J to measure synaptic density. Parametric distribution volume ratio (DVR) images using equilibrium methods were generated from dynamic images. For [18F]FPEB PET, DVR was calculated using equilibrium methods and a cerebellum reference region. For [11C]UCB-J PET, DVR was calculated with a simplified reference tissue model - 2 and a whole cerebellum reference region. RESULTS A strong positive correlation between mGluR5 and synaptic density was present in the hippocampus for participants with AD (r = 0.81, p < 0.001) and in the CN group (r = 0.74, p = 0.005). In the entorhinal cortex, there was a strong positive correlation between mGluR5 and synaptic density in the AD group (r = 0.85, p < 0.001), but a weaker non-significant correlation in the CN group (r = 0.36, p = 0.245). Exploratory analyses indicated more widespread significant positive correlations between synaptic density and mGluR5 within regions, as well as significant positive correlations between synaptic density in the temporal lobe and mGluR5 across a broader set of regions commonly affected by AD. CONCLUSIONS Our findings suggest that mGluR5 reduction in AD is closely linked to synaptic loss. Longitudinal studies are needed to clarify causality, deepen understanding of AD pathogenesis, and aid in developing novel biomarkers and treatments.
Collapse
Affiliation(s)
- Elaheh Salardini
- Alzheimer's Disease Research Unit, Yale University School of Medicine, One Church Street, 8 Floor, New Haven, CT, 06510, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Ryan S O'Dell
- Alzheimer's Disease Research Unit, Yale University School of Medicine, One Church Street, 8 Floor, New Haven, CT, 06510, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Em Tchorz
- Alzheimer's Disease Research Unit, Yale University School of Medicine, One Church Street, 8 Floor, New Haven, CT, 06510, USA
| | - Nabeel B Nabulsi
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Christopher H van Dyck
- Alzheimer's Disease Research Unit, Yale University School of Medicine, One Church Street, 8 Floor, New Haven, CT, 06510, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Adam P Mecca
- Alzheimer's Disease Research Unit, Yale University School of Medicine, One Church Street, 8 Floor, New Haven, CT, 06510, USA.
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
Kim JW, Byun MS, Yi D, Jung JH, Kong N, Chang YY, Jung G, Ahn H, Lee JY, Kang KM, Sohn CH, Lee YS, Kim YK, Lee DY. Daily fluid intake and brain amyloid deposition: A cohort study. J Alzheimers Dis 2025; 104:138-149. [PMID: 39980438 PMCID: PMC11934770 DOI: 10.1177/13872877251314176] [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: 02/22/2025]
Abstract
BackgroundLittle information is yet available for the association between daily water intake, a modifiable lifestyle factor, and Alzheimer's disease (AD) pathology and cerebrovascular injury in the living human brain.ObjectiveOur aim was to explore the correlation between daily fluid intake and in vivo AD pathologies (i.e., amyloid-β (Aβ) and tau) and cerebrovascular injury.Methods287 cognitively normal (CN) older adults completed extensive clinical assessments, daily fluid intake evaluations, and multimodal brain imaging at both the initial baseline and the subsequent 2-year follow-up.ResultsLow daily fluid intake was significantly associated with a higher level or a more rapid increase of Aβ deposition, especially in apolipoprotein E4 negative individuals. Meanwhile, low daily fluid intake was cross-sectionally related with cerebrovascular injury.ConclusionsOur findings suggest that high daily fluid intake is associated with decreased brain amyloid deposition, indicating that sufficient daily fluid intake may be helpful for prevention of AD.
Collapse
Affiliation(s)
- Jee Wook Kim
- Department of Neuropsychiatry, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Gyeonggi, Republic of Korea
- Department of Psychiatry, Hallym University College of Medicine, Chuncheon, Gangwon, Republic of Korea
| | - Min Soo Byun
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, Republic of Korea
| | - Joon Hyung Jung
- Department of Psychiatry, Chungbuk National University Hospital, Cheongju, Republic of Korea
| | - Nayeong Kong
- Department of Psychiatry, Keimyung University Dongsan Hospital, Daegu, Republic of Korea
| | - Yoon Young Chang
- Department of Psychiatry, Inje University Sanggye Paik Hospital, Seoul, Republic of Korea
| | - Gijung Jung
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, Republic of Korea
| | - Hyejin Ahn
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, Republic of Korea
| | - Jun-Young Lee
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Neuropsychiatry, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea
| | - Dong Young Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
6
|
Keum M, Byun MS, Yi D, Ahn H, Jung G, Jung JH, Kong N, Lee JY, Kim YK, Lee YS, Kang KM, Sohn CH, Lee DY. The Use of Antihypertensive Medication and In Vivo Alzheimer's Disease Pathology. Ann Neurol 2025. [PMID: 39960250 DOI: 10.1002/ana.27204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 03/05/2025]
Abstract
OBJECTIVE We investigated whether the use of antihypertensive medication (AHM) is associated with in vivo Alzheimer's Disease (AD) pathologies in older adults with hypertension and examined if the effect differs by drug-class and blood-brain barrier (BBB) permeability of the drug. METHODS This cross-sectional study recruited participants from the Korean Brain Aging Study for the Early Diagnosis and Prevention of Alzheimer's Disease. Participants comprised both cognitively normal and impaired older adults diagnosed with hypertension (n = 408). All participants underwent comprehensive clinical assessment and [11C] Pittsburgh Compound B positron emission tomography (PET) for measurement of cerebral β-amyloid (Aβ) deposition. Additionally, a subset of participants (n = 120) was subjected to [18F] AV-1451 PET to assess tau deposition. RESULTS The AHM group (n = 227) exhibited significantly lower Aβ deposition (B [SE] = -0.104 [0.037], p = 0.006) compared to the non-AHM group (n = 181), even after controlling for age, sex, apolipoprotein E ε4-positivity, vascular risk factors, and mean arterial blood pressure. Further analysis by AHM class showed an association between the use of renin-angiotensin system inhibitors (RASi) and less Aβ deposition (B [SE] = -0.143[0.049], p = 0.004). No significant relationships were observed between the use of BBB-permeable AHM and Aβ deposition. Additionally, associations between AHM use and tau deposition did not reach statistical significance. INTERPRETATION Our findings suggest that AHM use may be associated with lower Aβ burden in older adults with hypertension. Further studies exploring the underlying mechanism, particularly related to RASi, may provide insights into new therapeutic targets for AD. ANN NEUROL 2025.
Collapse
Affiliation(s)
- Musung Keum
- Department of Neuropsychiatry, Hallym University Dongtan Sacred Heart Hospital, Gyeonggi, Republic of Korea
| | - Min Soo Byun
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
- Convergence Research Center for Dementia, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Hyejin Ahn
- Interdisciplinary Program of Cognitive Science, Seoul National University College of Humanities, Seoul, South Korea
| | - Gijung Jung
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Joon Hyung Jung
- Department of Psychiatry, Chungbuk National University Hospital, Cheongju, Republic of Korea
| | - Nayeong Kong
- Department of Psychiatry, Keimyung University Dongsan Hospital, Daegu, Republic of Korea
| | - Jun-Young Lee
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Psychiatry, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Dong Young Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
- Convergence Research Center for Dementia, Seoul National University Medical Research Center, Seoul, Republic of Korea
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
- Interdisciplinary Program of Cognitive Science, Seoul National University College of Humanities, Seoul, South Korea
| |
Collapse
|
7
|
Grahl MVC, Hohl KS, Smaniotto T, Carlini CR. Microbial Trojan Horses: Virulence Factors as Key Players in Neurodegenerative Diseases. Molecules 2025; 30:687. [PMID: 39942791 PMCID: PMC11820544 DOI: 10.3390/molecules30030687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/01/2025] [Accepted: 02/01/2025] [Indexed: 02/16/2025] Open
Abstract
Changes in population demographics indicate that the elderly population will reach 2.1 billion worldwide by 2050. In parallel, there will be an increase in neurodegenerative diseases such as Alzheimer's and Parkinson's. This review explores dysbiosis occurring in these pathologies and how virulence factors contribute to the worsening or development of clinical conditions, and it summarizes existing and potential ways to combat microorganisms related to these diseases. Microbiota imbalances can contribute to the progression of neurodegenerative diseases by increasing intestinal permeability, exchanging information through innervation, and even acting as a Trojan horse affecting immune cells. The microorganisms of the microbiota produce virulence factors to protect themselves from host defenses, many of which contribute to neurodegenerative diseases. These virulence factors are expressed according to the genetic composition of each microorganism, leading to a wide range of factors to be considered. Among the main virulence factors are LPS, urease, curli proteins, amyloidogenic proteins, VacA, and CagA. These factors can also be packed into bacterial outer membrane vesicles, which transport proteins, RNA, and DNA, enabling distal communication that impacts various diseases, including Alzheimer's and Parkinson's.
Collapse
Affiliation(s)
- Matheus V. C. Grahl
- Graduate Program in Medicine and Health Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90619-900, RS, Brazil
| | - Kelvin Siqueira Hohl
- Graduate Program in Biochemistry, Institute of Health Basic Sciences, Federal University of Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil; (K.S.H.); (T.S.)
| | - Thiago Smaniotto
- Graduate Program in Biochemistry, Institute of Health Basic Sciences, Federal University of Rio Grande do Sul, Porto Alegre 90035-003, RS, Brazil; (K.S.H.); (T.S.)
| | - Célia R. Carlini
- Center of Biotechnology, Department of Biochemistry, Federal University of Rio Grande do Sul, Porto Alegre 91501-970, RS, Brazil
- Graduate Program of Biosciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050-170, RS, Brazil
| |
Collapse
|
8
|
Yi D, Byun MS, Park J, Kim J, Jung G, Ahn H, Lee J, Lee Y, Kim YK, Kang KM, Sohn C, Liu S, Huang Y, Saykin AJ, Lee DY, Nho K. Tau pathway-based gene analysis on PET identifies CLU and FYN in a Korean cohort. Alzheimers Dement 2025; 21:e14416. [PMID: 39625110 PMCID: PMC11848168 DOI: 10.1002/alz.14416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/25/2024] [Accepted: 10/30/2024] [Indexed: 12/13/2024]
Abstract
INTRODUCTION The influence of genetic variation on tau protein aggregation, a key factor in Alzheimer's disease (AD), remains not fully understood. We aimed to identify novel genes associated with brain tau deposition using pathway-based candidate gene association analysis in a Korean cohort. METHODS We analyzed data for 146 older adults from the well-established Korean AD continuum cohort (Korean Brain Aging Study for the Early Diagnosis and Prediction of Alzheimer's Disease; KBASE). Fifteen candidate genes related to both tau pathways and AD were selected. Association analyses were performed using PLINK: A tool set for whole-genome association and population-based linkage analyses (PLINK) on tau deposition measured by 18F-AV-1451 positron emission tomography (PET) scans, with additional voxel-wise analysis conducted using Statistical Parametric Mapping 12 (SPM12). RESULTS CLU and FYN were significantly associated with tau deposition, with the most significant single-nucleotide polymorphisms (SNPs) being rs149413552 and rs57650567, respectively. These SNPs were linked to increased tau across key brain regions and showed additive effects with apolipoprotein E (APOE) ε4. DISCUSSION CLU and FYN may play specific roles in tau pathophysiology, offering potential targets for biomarkers and therapies. HIGHLIGHTS Gene-based analysis identified CLU and FYN as associated with tau deposition on positron emission tomography (PET). CLU rs149413552 and FYN rs57650567 were associated with brain tau deposition. rs149413552 and rs57650567 were associated with structural brain atrophy. CLU rs149413552 was associated with immediate verbal memory. CLU and FYN may play specific roles in tau pathophysiology.
Collapse
Affiliation(s)
- Dahyun Yi
- Institute of Human Behavioral MedicineMedical Research CenterSeoul National UniversitySeoulSouth Korea
| | - Min Soo Byun
- Department of NeuropsychiatrySeoul National University HospitalSeoulSouth Korea
- Department of PsychiatrySeoul National University College of MedicineSeoulSouth Korea
| | - Jong‐Ho Park
- Precision Medicine CenterSeoul National University Bundang HospitalSeongnam‐siGyeonggi‐doSouth Korea
| | - Jong‐Won Kim
- Department of Laboratory Medicine and GeneticsSamsung Medical CenterSungkyunkwan University School of MedicineGangnam‐guSeoulSouth Korea
| | - Gijung Jung
- Institute of Human Behavioral MedicineMedical Research CenterSeoul National UniversitySeoulSouth Korea
| | - Hyejin Ahn
- Interdisciplinary Program of Cognitive ScienceSeoul National University College of HumanitiesGwanak‐guSeoulSouth Korea
| | - Jun‐Young Lee
- Department of PsychiatrySeoul National University Boramae Medical Center, Dongjak‐guSeoulSouth Korea
| | - Yun‐Sang Lee
- Department of Nuclear MedicineSeoul National University College of MedicineJongro‐guSeoulSouth Korea
| | - Yu Kyeong Kim
- Department of Nuclear MedicineSeoul National University Boramae Medical Center, Dongjak‐guSeoulSouth Korea
| | - Koung Mi Kang
- Department of RadiologySeoul National University Hospital, Jongro‐guSeoulSouth Korea
- Department of RadiologySeoul National University College of Medicine, Jongro‐guSeoulSouth Korea
| | - Chul‐Ho Sohn
- Department of RadiologySeoul National University Hospital, Jongro‐guSeoulSouth Korea
| | - Shiwei Liu
- Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Center for NeuroimagingDepartment of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Yen‐Ning Huang
- Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Center for NeuroimagingDepartment of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Andrew J. Saykin
- Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Center for NeuroimagingDepartment of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Dong Young Lee
- Institute of Human Behavioral MedicineMedical Research CenterSeoul National UniversitySeoulSouth Korea
- Department of NeuropsychiatrySeoul National University HospitalSeoulSouth Korea
- Department of PsychiatrySeoul National University College of MedicineSeoulSouth Korea
| | - Kwangsik Nho
- Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Center for NeuroimagingDepartment of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | | |
Collapse
|
9
|
Ko K, Yi D, Byun MS, Jung JH, Kong N, Jung G, Ahn H, Chang YY, Keum M, Lee J, Lee Y, Kim YK, Lee DY. Moderation of midlife cognitive activity on tau-related cognitive impairment. Alzheimers Dement 2025; 21:e14606. [PMID: 39988958 PMCID: PMC11847999 DOI: 10.1002/alz.14606] [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: 08/29/2024] [Revised: 12/24/2024] [Accepted: 01/17/2025] [Indexed: 02/25/2025]
Abstract
INTRODUCTION We investigated the moderating effects of midlife and late-life cognitive activity (CA) on the relationship between tau pathology and both cognition and cognitive decline. METHODS Eighty-nine non-demented older adults from a Korean cohort underwent comprehensive evaluations, including CA assessments and tau neuroimaging at baseline, and Mini-Mental State Examination (MMSE) at baseline and the 2-year follow-up. RESULTS Greater midlife CA was associated with higher MMSE scores in a given amount of tau pathology, whereas higher levels of midlife CA were associated with faster tau-related decline in MMSE scores, particularly in individuals with mild cognitive impairment. Late-life CA did not exhibit any interaction with tau on either MMSE scores or their 2-year change. DISCUSSION Greater midlife CA is generally associated with better cognitive performance despite the presence of tau pathology. However, paradoxically, increased midlife CA appears to be linked to a more rapid tau-related cognitive decline in already cognitively impaired individuals. HIGHLIGHTS Greater midlife cognitive activity (CA) was generally associated with better cognitive performance in a given amount of tau pathology. Paradoxically, higher levels of midlife CA were related to a more rapid tau-related cognitive decline in already cognitively impaired individuals. Late-life CA did not exhibit any moderation effect on the association between tau and cognitive performance or decline.
Collapse
Grants
- U01AG072177 Ministry of Science, ICT, and Future Planning, Republic of Korea
- 27303C0140 NIEHS NIH HHS
- RS-2022-00165636 Ministry of Science, ICT, and Future Planning, Republic of Korea
- NRF-2014M3C7A1046042 Ministry of Science, ICT, and Future Planning, Republic of Korea
- HU23C0140 Ministry of Science, ICT, and Future Planning, Republic of Korea
- HI18C0630 Ministry of Science, ICT, and Future Planning, Republic of Korea
- U01 AG072177 NIA NIH HHS
- HI19C0149 Ministry of Science, ICT, and Future Planning, Republic of Korea
Collapse
Affiliation(s)
- Kang Ko
- Department of PsychiatrySeoul National University College of MedicineSeoulRepublic of Korea
- Department of PsychiatryNational Health Insurance Service Ilsan HospitalGoyangRepublic of Korea
| | - Dahyun Yi
- Institute of Human Behavioral MedicineMedical Research CenterSeoul National UniversitySeoulRepublic of Korea
| | - Min Soo Byun
- Department of PsychiatrySeoul National University College of MedicineSeoulRepublic of Korea
- Department of NeuropsychiatrySeoul National University HospitalSeoulRepublic of Korea
| | - Joon Hyung Jung
- Department of PsychiatryChungbuk National University HospitalCheongjuRepublic of Korea
| | - Nayeong Kong
- Department of PsychiatryKeimyung University Dongsan HospitalDaeguRepublic of Korea
| | - Gijung Jung
- Department of NeuropsychiatrySeoul National University HospitalSeoulRepublic of Korea
| | - Hyejin Ahn
- Department of NeuropsychiatrySeoul National University HospitalSeoulRepublic of Korea
| | - Yoon Young Chang
- Department of PsychiatryInje University Sanggye Paik HospitalSeoulRepublic of Korea
| | - Musung Keum
- Department of NeuropsychiatrySeoul National University HospitalSeoulRepublic of Korea
| | - Jun‐Young Lee
- Department of PsychiatrySeoul National University College of MedicineSeoulRepublic of Korea
- Department of NeuropsychiatrySMG‐SNU Boramae Medical CenterSeoulRepublic of Korea
| | - Yun‐Sang Lee
- Department of Nuclear MedicineSeoul National University College of MedicineSeoulRepublic of Korea
| | - Yu Kyeong Kim
- Department of Nuclear MedicineSMG‐SNU Boramae Medical CenterSeoulRepublic of Korea
| | - Dong Young Lee
- Department of PsychiatrySeoul National University College of MedicineSeoulRepublic of Korea
- Institute of Human Behavioral MedicineMedical Research CenterSeoul National UniversitySeoulRepublic of Korea
- Department of NeuropsychiatrySeoul National University HospitalSeoulRepublic of Korea
| | | |
Collapse
|
10
|
Biel D, Suárez-Calvet M, Dewenter A, Steward A, Roemer SN, Dehsarvi A, Zhu Z, Pescoller J, Frontzkowski L, Kreuzer A, Haass C, Schöll M, Brendel M, Franzmeier N. Female sex is linked to a stronger association between sTREM2 and CSF p-tau in Alzheimer's disease. EMBO Mol Med 2025; 17:235-248. [PMID: 39794447 PMCID: PMC11822105 DOI: 10.1038/s44321-024-00190-3] [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: 08/02/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 01/13/2025] Open
Abstract
In Alzheimer's disease (AD), Aβ triggers p-tau secretion, which drives tau aggregation. Therefore, it is critical to characterize modulators of Aβ-related p-tau increases which may alter AD trajectories. Here, we assessed whether factors known to alter tau levels in AD modulate the association between fibrillar Aβ and secreted p-tau181 determined in the cerebrospinal fluid (CSF). To assess potentially modulating effects of female sex, younger age, and ApoE4, we included 322 ADNI participants with cross-sectional/longitudinal p-tau181. To determine effects of microglial activation on p-tau181, we included 454 subjects with cross-sectional CSF sTREM2. Running ANCOVAs for nominal and linear regressions for metric variables, we found that women had higher Aβ-related p-tau181 levels. Higher sTREM2 was associated with elevated p-tau181, with stronger associations in women. Similarly, ApoE4 was related to higher p-tau181 levels and faster p-tau181 increases, with stronger effects in female ApoE4 carriers. Our results show that sex alone modulates the Aβ to p-tau axis, where women show higher Aβ-dependent p-tau secretion, potentially driven by elevated sTREM2-related microglial activation and stronger effects of ApoE4 carriership in women.
Collapse
Affiliation(s)
- Davina Biel
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany.
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
- Servei de Neurologia, Hospital del Mar, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Anna Dewenter
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Anna Steward
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Sebastian N Roemer
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Department of Neurology, University Hospital, LMU Munich, Munich, Germany
| | - Amir Dehsarvi
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Zeyu Zhu
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Julia Pescoller
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Lukas Frontzkowski
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Annika Kreuzer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Chair of Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Munich, Germany
| | - Michael Schöll
- University of Gothenburg, The Sahlgrenska Academy, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Mölndal and Gothenburg, Sweden
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- University of Gothenburg, The Sahlgrenska Academy, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Mölndal and Gothenburg, Sweden
| |
Collapse
|
11
|
Yi D, Byun MS, Jung JH, Jung G, Ahn H, Chang YY, Keum M, Lee J, Lee Y, Kim YK, Kang KM, Sohn C, Risacher SL, Saykin AJ, Lee DY. Locus coeruleus tau is linked to successive cortical tau accumulation. Alzheimers Dement 2025; 21:e14426. [PMID: 39641328 PMCID: PMC11848382 DOI: 10.1002/alz.14426] [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: 08/30/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION We investigated the hypothesis that tau burden in the locus coeruleus (LC) correlates with tau accumulation in cortical regions according to the Braak stages and examined whether the relationships differed according to cortical amyloid beta (Aβ) deposition. METHODS One hundred and seventy well-characterized participants from an ongoing cohort were included. High-resolution T1, tau positron emission tomography (PET), and amyloid PET were obtained. RESULTS LC tau burden was significantly linked to global tau in neocortical regions, as was tau in both early Braak stage (stage I/II) and later Braak stage areas. This relationship was significant only in Aβ-positive individuals. While LC tau did not directly impact memory, it was indirectly associated with delayed memory through mediation or moderation pathways. DISCUSSION The findings from living human brains support the idea that LC tau closely relates to subsequent cortical tau accumulation, particularly among individuals with pathological Aβ accumulation, and identify LC tau burden as a promising indicator of cognitive trajectories of AD. HIGHLIGHTS Tau burden in the LC was significantly associated with cortical tau accumulation. Tau burden in SN or PPN showed no association with cortical tau accumulation. LC tau burden was serially associated with Braak stages. The tau-LC and cortical tau relationship was significant only in the Aβ-positive group. Cortical amyloid's impact on memory worsens with higher tau accumulation in the LC.
Collapse
Affiliation(s)
- Dahyun Yi
- Institute of Human Behavioral MedicineMedical Research CenterSeoul National UniversitySeoulSeoulRepublic of Korea
| | - Min Soo Byun
- Department of NeuropsychiatrySeoul National University HospitalSeoulSeoulRepublic of Korea
- Department of PsychiatrySeoul National University College of MedicineSeoulSeoulRepublic of Korea
| | - Joon Hyung Jung
- Department of PsychiatryChungbuk National University HospitalCheongju‐siRepublic of Korea
| | - Gijung Jung
- Institute of Human Behavioral MedicineMedical Research CenterSeoul National UniversitySeoulSeoulRepublic of Korea
| | - Hyejin Ahn
- Interdisciplinary Program of Cognitive ScienceSeoul National University College of HumanitiesSeoulSeoulRepublic of Korea
| | - Yoon Young Chang
- Inje University Sanggye Paik HospitalSeoulSeoulRepublic of Korea
| | - Musung Keum
- Department of NeuropsychiatrySoonchunhyang University HospitalBucheon‐siGyeonggi‐doRepublic of Korea
| | - Jun‐Young Lee
- Department of PsychiatrySeoul National University Boramae Medical CenterSeoulSeoulRepublic of Korea
| | - Yun‐Sang Lee
- Department of Nuclear MedicineSeoul National University College of MedicineSeoulSeoulRepublic of Korea
| | - Yu Kyeong Kim
- Department of Nuclear MedicineSeoul National University Boramae Medical CenterSeoulSeoulRepublic of Korea
| | - Koung Mi Kang
- Department of RadiologySeoul National University HospitalSeoulSeoulRepublic of Korea
- Department of RadiologySeoul National University College of MedicineSeoulSeoulRepublic of Korea
| | - Chul‐Ho Sohn
- Department of RadiologySeoul National University HospitalSeoulSeoulRepublic of Korea
- Department of RadiologySeoul National University College of MedicineSeoulSeoulRepublic of Korea
| | - Shannon L. Risacher
- Indiana Alzheimer's Disease Research CenterIndianapolisIndianaUSA
- Center for NeuroimagingDepartment of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
| | - Andrew J. Saykin
- Indiana Alzheimer's Disease Research CenterIndianapolisIndianaUSA
- Center for NeuroimagingDepartment of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Network Science InstituteIndiana UniversityBloomingtonIndianaUSA
| | - Dong Young Lee
- Institute of Human Behavioral MedicineMedical Research CenterSeoul National UniversitySeoulSeoulRepublic of Korea
- Department of NeuropsychiatrySeoul National University HospitalSeoulSeoulRepublic of Korea
- Department of PsychiatrySeoul National University College of MedicineSeoulSeoulRepublic of Korea
| | | |
Collapse
|
12
|
Huang S, Nunez J, Toresco DL, Wen C, Slotabec L, Wang H, Zhang H, Rouhi N, Adenawoola MI, Li J. Alterations in the inflammatory homeostasis of aging-related cardiac dysfunction and Alzheimer's diseases. FASEB J 2025; 39:e70303. [PMID: 39758048 DOI: 10.1096/fj.202402725rr] [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: 11/05/2024] [Revised: 12/20/2024] [Accepted: 12/26/2024] [Indexed: 01/07/2025]
Abstract
Alzheimer's disease (AD) is well known among the elderly and has a profound impact on both patients and their families. Increasing research indicates that AD is a systemic disease, with a strong connection to cardiovascular disease. They share common genetic factors, such as mutations in the presenilin (PS1 and PS2) and the apolipoprotein E (APOE) genes. Cardiovascular conditions can lead to reduced cerebral blood flow and increased oxidative stress. These factors contribute to the accumulation of Aβ plaques and the formation of abnormal tau protein tangles, which are both key pathological features of AD. Additionally, Aβ deposits and abnormal protein responses have been observed in cardiomyocytes as well as in peripheral tissues. The toxic Aβ deposition intensifies damage to the microvascular structure associated with blood-brain barrier disruption and the initiation of neuroinflammation, which may accelerate the onset of neurocognitive deficits and cardiovascular dysfunction. Thus, we discuss the main mechanisms linking AD and cardiac dysfunction to enhance our understanding of these conditions. Ultimately, insights into the brain-heart axis may help us develop effective treatment strategies in the future.
Collapse
Affiliation(s)
- Shuli Huang
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Jeremiah Nunez
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi, USA
| | - Dai Lan Toresco
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi, USA
| | - Changhong Wen
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi, USA
| | - Lily Slotabec
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi, USA
| | - Hao Wang
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Haibei Zhang
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Nadiyeh Rouhi
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Michael I Adenawoola
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Ji Li
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
13
|
Mares J, Kumar G, Sharma A, Emrani S, McIntire LB, Guo J, Menon V, Nuriel T. APOE ε4-associated heterogeneity of neuroimaging biomarkers across the Alzheimer's disease continuum. Alzheimers Dement 2025; 21:e14392. [PMID: 39575672 PMCID: PMC11775459 DOI: 10.1002/alz.14392] [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: 05/06/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 01/30/2025]
Abstract
INTRODUCTION While the role of apolipoprotein E (APOE) ε4 in Alzheimer's disease (AD) susceptibility has been studied extensively, much less is known about the differences in disease presentation in APOE ε4 carriers versus non-carriers. METHODS To help elucidate these differences, we performed a broad analysis comparing the regional levels of six different neuroimaging biomarkers in the brains of APOE ε4 carriers versus non-carriers who participated in the Alzheimer's Disease Neuroimaging Initiative (ADNI). RESULTS We observed significant APOE ε4-associated heterogeneity in regional amyloid beta deposition, tau accumulation, glucose uptake, brain volume, cerebral blood flow, and white matter hyperintensities within each AD diagnostic group. We also observed important APOE ε4-associated differences in cognitively unimpaired individuals who converted to mild cognitive impairment/AD versus those who did not convert. DISCUSSION This observed heterogeneity in neuroimaging biomarkers between APOE ε4 carriers versus non-carriers may have important implications regarding the prevention, diagnosis, and treatment of AD in different subpopulations. HIGHLIGHTS An extensive study was performed on the apolipoprotein E (APOE) ε4-associated heterogeneity in neuroimaging biomarkers from the Alzheimer's Disease Neuroimaging Initiative. Robust APOE ε4-associated increases in amyloid beta (Aβ) deposition throughout the brain, in every diagnostic group, were observed. APOE ε4-associated increases in tau pathology, decreases in glucose uptake, and increases in brain atrophy, which expand in regional scope and magnitude with disease progression, were observed. Significant sex- and age-related differences in APOE ε4-associated neuroimaging biomarker heterogeneity, with overall increases in pathological presentation in female APOE ε4 carriers, were observed. Regional differences in Aβ deposition, tau accumulation, glucose uptake, ventricle size, and white matter hyperintensities were observed in cognitively normal participants who converted to mild cognitive impairment/Alzheimer's disease, which may hold potential predictive value.
Collapse
Affiliation(s)
- Jason Mares
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- Department of NeurologyColumbia UniversityNew YorkNew YorkUSA
| | - Gautam Kumar
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- Department of Pathology and Cell BiologyColumbia UniversityNew YorkNew YorkUSA
- Department of NeurobiologyUniversity of MarylandBaltimoreMarylandUSA
| | - Anurag Sharma
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- Department of Pathology and Cell BiologyColumbia UniversityNew YorkNew YorkUSA
| | - Sheina Emrani
- Department of NeurologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Laura Beth McIntire
- Lipidomics and Biomarker Discovery LabBrain Health Imaging InstituteDepartment of RadiologyWeill Cornell MedicineNew YorkNew YorkUSA
| | - Jia Guo
- Department of PsychiatryColumbia UniversityNew YorkNew YorkUSA
- Zuckerman InstituteColumbia UniversityNew YorkNew YorkUSA
| | - Vilas Menon
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- Department of NeurologyColumbia UniversityNew YorkNew YorkUSA
| | - Tal Nuriel
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- Department of Pathology and Cell BiologyColumbia UniversityNew YorkNew YorkUSA
| | | |
Collapse
|
14
|
Gol Mohammad Pour Afrakoti L, Daneshpour Moghadam S, Hadinezhad P. Alzheimer's disease and the immune system: A comprehensive overview with a focus on B cells, humoral immunity, and immunotherapy. J Alzheimers Dis Rep 2025; 9:25424823251329188. [PMID: 40297057 PMCID: PMC12035277 DOI: 10.1177/25424823251329188] [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: 05/08/2024] [Accepted: 02/11/2025] [Indexed: 04/30/2025] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder and the major cause of dementia. Amyloid-β (Aβ) and tau aggregation, mitochondrial dysfunction, and microglial dysregulation are key contributors to AD pathogenesis. Impairments in the blood-brain barrier have unveiled the contribution of the immune system, particularly B cells, in AD pathology. B cells, a crucial component of adaptive immunity, exhibit diverse functions, including antigen presentation and antibody production. While their role in neuroinflammatory disorders has been well-documented, their specific function in AD lacks adequate data. This review examines the dual role of the B cells and humoral immunity in modulating brain inflammation in AD and explores recent advancements in passive and active immunotherapeutic strategies targeting AD pathobiology. We summarize preclinical and clinical studies investigating B cell frequency, altered antibody levels, and their implications in neuroinflammation and immunotherapy. Notably, B cells demonstrate protective and pathological roles in AD, influencing neurodegeneration through antibody-mediated clearance of toxic aggregates and inflammatory activation inflammation. Passive immunotherapies targeting Aβ have shown potential in reducing amyloid plaques, while active immunotherapies are emerging as promising strategies, requiring further validation. Understanding the interplay between B cells, humoral immunity, microglia, and mitochondrial dysfunction is critical to unraveling AD pathogenesis. Their dual nature in disease progression underscores the need for precise therapeutic interventions to optimize immunotherapy outcomes and mitigate neuroinflammation effectively.
Collapse
Affiliation(s)
| | - Sanam Daneshpour Moghadam
- Department of Diagnostic and Public Health, School of Biotechnology, University of Verona, Verona, Italy
| | - Pezhman Hadinezhad
- Cognitive Neurology, Dementia and Neuropsychiatry Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Tripathi S, Sharma Y, Kumar D. Unraveling APOE4's Role in Alzheimer's Disease: Pathologies and Therapeutic Strategies. Curr Protein Pept Sci 2025; 26:259-281. [PMID: 39722484 DOI: 10.2174/0113892037326839241014054430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/13/2024] [Accepted: 08/30/2024] [Indexed: 12/28/2024]
Abstract
Alzheimer's disease (AD), the most common kind of dementia worldwide, is characterized by elevated levels of the amyloid-β (Aβ) peptide and hyperphosphorylated tau protein in the neurons. The complexity of AD makes the development of treatments infamously challenging. Apolipoprotein E (APOE) genes's ε4 allele is one of the main genetic risk factors for AD. While the APOE gene's ε4 allele considerably increases the chance of developing AD, the ε2 allele is protective compared to the prevalent ε3 variant. It is fiercely discussed how APOE affects the development and course of disease since it has a variety of activities that influence both neuronal and non-neuronal cells. ApoE4 contributes to the formation of tau tangles, deposition of Aβ, neuroinflammation, and other processes. Four decades of research have provided a significant understanding of the structure of APOE and how this may affect the neuropathology and pathogenesis of AD. APOE is a crucial lipid transporter essential for the growth of the central nervous system (CNS), upkeep, and repair. The mechanisms by which APOE contributes to the pathophysiology of AD are still up for discussion, though. Evidence suggests that APOE affects the brain's clearance and deposition of Aβ. Additionally, APOE has Aβ-independent pathways in AD, which has led to the identification of new functions for APOE, including mitochondrial dysfunction. This study summarizes important studies that describe how APOE4 affects well-known AD pathologies, including tau pathology, Aβ, neuroinflammation, and dysfunction of neural networks. This study also envisions some of the therapeutic approaches being used to target APOE4 in the hopes of preventing or treating AD.
Collapse
Affiliation(s)
- Siddhant Tripathi
- Department of Pharmaceutical Chemistry, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, 411038, India
| | - Yashika Sharma
- Department of Pharmaceutical Chemistry, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, 411038, India
| | - Dileep Kumar
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| |
Collapse
|
16
|
Jung JH, Byun MS, Yi D, Ahn H, Lee JH, Lee JS, Lee HS, Lee JY, Kim YK, Lee YS, Kang KM, Sohn CH, Lee DY. Telomere length, in vivo Alzheimer's disease pathologies and cognitive decline in older adults. J Neurol Neurosurg Psychiatry 2024:jnnp-2024-334314. [PMID: 39667908 DOI: 10.1136/jnnp-2024-334314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 11/17/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND Whether telomere length (TL), an indicator of biological ageing, reflects Alzheimer's disease (AD)-related neuropathological change remains unclear. We investigated the relationships between TL, in vivo AD pathologies, including cerebral beta-amyloid and tau deposition, and cognitive outcomes in older adults. METHODS A total of 458 older adults were included, encompassing both cognitively normal (CN) individuals and those cognitively impaired (CI), with the CI group consisting of individuals with mild cognitive impairment or AD dementia. All participants underwent clinical and neuropsychological assessments, amyloid positron emission tomography (PET) scan and DNA extraction for measuring TL at baseline. A subset of participants (n=140) underwent tau PET scan. At follow-up, the participants underwent neuropsychological assessments annually for up to 4 years. RESULTS Overall, longer TL was associated with greater brain tau deposition (B=0.139, 95% CI 0.040, 0.238) and a faster decline in global cognition (B = - 0.371, 95% CI - 0.720, -0.023). In the subgroup analysis, the association between longer TL and greater in vivo AD pathologies, as well as faster cognitive decline, was observed particularly in the CI group. Mediation analysis suggested that longer TL was associated with cognitive decline through increased tau deposition in the CI group. CONCLUSION Our finding suggests that older adults with relatively longer TL, particularly in the CI group, may have greater in vivo AD pathologies and experience more rapid cognitive decline, potentially mediated by brain tau deposition. Further studies are necessary to elucidate the biological links underlying these associations.
Collapse
Affiliation(s)
- Joon Hyung Jung
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
- Department of Psychiatry, Chungbuk National University Hospital, Cheongju, Korea (the Republic of)
| | - Min Soo Byun
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Korea (the Republic of)
- Convergence Research Center for Dementia, Seoul National University Medical Research Center, Seoul, Korea (the Republic of)
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Seoul National University Medical Research Center, Seoul, Korea (the Republic of)
| | - Hyejin Ahn
- Interdisciplinary Program of Cognitive Neuroscience, Seoul National University College of Humanities, Seoul, Korea (the Republic of)
| | - Jun Ho Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Korea (the Republic of)
| | - Jang-Seok Lee
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea (the Republic of)
| | - Hyun-Seob Lee
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea (the Republic of)
| | - Jun-Young Lee
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
- Department of Neuropsychiatry, Seoul Metropolitan Government-Seoul National University (SMG-SNU) Boramae Medical Center, Seoul, Korea (the Republic of)
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, Seoul Metropolitan Government-Seoul National University (SMG-SNU) Boramae Medical Center, Seoul, Korea (the Republic of)
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, Seoul, Korea (the Republic of)
- Department of Radiology, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, Seoul, Korea (the Republic of)
- Department of Radiology, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
| | - Dong Young Lee
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Korea (the Republic of)
- Convergence Research Center for Dementia, Seoul National University Medical Research Center, Seoul, Korea (the Republic of)
- Institute of Human Behavioral Medicine, Seoul National University Medical Research Center, Seoul, Korea (the Republic of)
- Interdisciplinary Program of Cognitive Neuroscience, Seoul National University College of Humanities, Seoul, Korea (the Republic of)
| |
Collapse
|
17
|
Jang H, Chun MY, Yun J, Kim JP, Kang SH, Kim HJ, Na DL, Lee EH, Shin D, Ham H, Gu Y, Kim CH, Woo SY, Seo SW. Distinct Cognitive Trajectories According to Amyloid Positivity in Non-Alzheimer Disease Dementias. Clin Nucl Med 2024; 49:1073-1078. [PMID: 39385364 DOI: 10.1097/rlu.0000000000005457] [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: 10/12/2024]
Abstract
BACKGROUND The clinical effects of β-amyloid positivity (Aβ+) on copathologies in various dementias remain relatively underexamined. Thus, the present study was conducted to investigate the prevalence and clinical effects of Aβ+ in subcortical vascular cognitive impairment (SVCI) and frontotemporal dementia (FTD). PATIENTS AND METHODS We enrolled SVCI (n = 583), FTD (n = 152), and cognitively unimpaired (CU) participants (n = 1,249) who underwent Aβ PET scans. The odds of having Aβ+ were subsequently compared among the diagnostic groups (CU, SVCI, and FTD) according to age and apolipoprotein E genotype. Additionally, a linear mixed-effects model was used to investigate the effects of Aβ+ on cognitive trajectories in SVCI and FTD. RESULTS Compared with CU, the SVCI group had a higher prevalence of Aβ+ in the 75 to 90 years age group (adjusted odds ratio, 1.97; 95% confidence interval, 1.36-2.85; P < 0.001), as well as within the apolipoprotein E ε3/ε3 group (adjusted odds ratio, 1.78; 95% confidence interval, 1.20-2.63; P = 0.001), whereas the FTD group showed no difference in Aβ+ prevalence. Aβ+ was associated with a worse cognitive trajectory in SVCI (adjusted β-coefficient = -0.6424; P < 0.001), but not in FTD. CONCLUSIONS These findings contribute to our understanding of Aβ biomarker traits in various dementias in Korea.
Collapse
Affiliation(s)
| | | | | | - Jun Pyo Kim
- From the Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sung Hoon Kang
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | | | | | - Eun Hye Lee
- From the Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Daeun Shin
- From the Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | | | | | - Chi-Hun Kim
- Department of Neurology, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Republic of Korea
| | - Sook-Young Woo
- Biomedical Statistics Center, Data Science Research Institute, Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | | |
Collapse
|
18
|
Byeon G, Byun MS, Yi D, Jung JH, Kong N, Chang Y, KEUM MUSUNG, Jung G, Ahn H, Lee JY, Kim YK, Kang KM, Sohn CH, Lee DY. Visual and Auditory Sensory Impairments Differentially Relate with Alzheimer's Pathology. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE : THE OFFICIAL SCIENTIFIC JOURNAL OF THE KOREAN COLLEGE OF NEUROPSYCHOPHARMACOLOGY 2024; 22:610-623. [PMID: 39420608 PMCID: PMC11494423 DOI: 10.9758/cpn.24.1169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 05/18/2024] [Accepted: 06/25/2024] [Indexed: 10/19/2024]
Abstract
Objective We intended to investigate the relationships between visual sensory impairment (VSI) or auditory sensory impairment (ASI) and brain pathological changes associated with cognitive decline in older adults. Methods We primarily tried to examine whether each sensory impairment is related to Alzheimer's disease (AD) pathology, specifically beta-amyloid (Aβ) deposition, through both cross-sectional and longitudinal approaches in cognitively unimpaired older adults. Self-report questionnaires on vision and hearing status were administered at the baseline. Neuroimaging scans including brain [11C] Pittsburgh Compound B PET and MRI, as well as clinical assessments, were performed at baseline and 2-year follow-up. Results Cross-sectional analyses showed that the VSI-positive group had significantly higher Aβ deposition than the VSI-negative group, whereas there was no significant association between ASI positivity and Aβ deposition. Longitudinal analyses revealed that VSI positivity at baseline was significantly associated with increased Aβ deposition over 2 years (β = 0.153, p = 0.025), although ASI positivity was not (β = 0.045, p = 0.518). VSI positivity at baseline was also significantly associated with greater atrophic changes in AD-related brain regions over the 2-year follow-up period (β = -0.207, p = 0.005), whereas ASI positivity was not (β = 0.024, p = 0.753). Neither VSI nor ASI positivity was related to cerebrovascular injury, as measured based on the white matter hyperintensity volume. Conclusion The findings suggest that VSI is probably related to AD-specific pathological changes, which possibly mediate the reported relationship between VSI and cognitive decline. In contrast, ASI appears not associated with AD pathologies but may contribute to cognitive decline via other mechanisms.
Collapse
Affiliation(s)
- Gihwan Byeon
- Department of Neuropsychiatry, Kangwon National University Hospital, Chuncheon, Korea
| | - Min Soo Byun
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Seoul National University Medical Research Center, Seoul, Korea
| | - Joon Hyung Jung
- Department of Psychiatry, Chungbuk National University Hospital, Cheongju, Korea
| | - Nayeong Kong
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea
| | - Yoonyoung Chang
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea
| | - MUSUNG KEUM
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea
| | - Gijung Jung
- Institute of Human Behavioral Medicine, Seoul National University Medical Research Center, Seoul, Korea
| | - Hyejin Ahn
- Institute of Human Behavioral Medicine, Seoul National University Medical Research Center, Seoul, Korea
| | - Jun-Young Lee
- Department of Neuropsychiatry, SMG-SNU Boramae Medical Center, Seoul, Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, Korea
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, Seoul, Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, Seoul, Korea
| | - Dong Young Lee
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Korea
- Institute of Human Behavioral Medicine, Seoul National University Medical Research Center, Seoul, Korea
| | | |
Collapse
|
19
|
Park J, Byun MS, Yi D, Ahn H, Jung JH, Kong N, Chang YY, Jung G, Lee JY, Kim YK, Lee YS, Kang KM, Sohn CH, Lee DY. The Moderating Effect of Serum Vitamin D on the Relationship between Beta-amyloid Deposition and Neurodegeneration. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE : THE OFFICIAL SCIENTIFIC JOURNAL OF THE KOREAN COLLEGE OF NEUROPSYCHOPHARMACOLOGY 2024; 22:646-654. [PMID: 39420611 PMCID: PMC11494430 DOI: 10.9758/cpn.24.1189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/01/2024] [Accepted: 06/09/2024] [Indexed: 10/19/2024]
Abstract
Objective Previous studies have reported that vitamin D deficiency increased the risk of Alzheimer's disease (AD) dementia in older adults. However, little is known about how vitamin D is involved in the pathophysiology of AD. Thus, this study aimed to examine the association and interaction of serum vitamin D levels with in vivo AD pathologies including cerebral beta-amyloid (Aβ) deposition and neurodegeneration in nondemented older adults. Methods 428 Nondemented older adults were recruited from the Korean Brain Aging Study for the Early Diagnosis and Prediction of Alzheimer's Disease, a prospective cohort that began in 2014. All participants underwent comprehensive clinical assessments, measurement of serum 25-hydroxyvitamin D (25[OH]D), and multimodal brain imaging including Pittsburgh compound B (PiB) positron emission tomography and magnetic resonance imaging. Global PiB deposition was measured for the Aβ biomarker. Intracranial volume-adjusted hippocampal volume (HVa) was used as a neurodegeneration biomarker. Results Overall, serum 25(OH)D level was not associated with either Aβ deposition or HVa after controlling for age, sex, apolipoprotein E ε4 positivity, and vascular risk factors. However, serum 25(OH)D level had a significant moderating effect on the association between Aβ and neurodegeneration, with lower serum 25(OH)D level significantly exacerbating cerebral Aβ-associated hippocampal volume loss (B = 34.612, p = 0.008). Conclusion Our findings indicate that lower serum vitamin D levels may contribute to AD by exacerbating Aβ-associated neurodegeneration in nondemented older adults. Further studies to explore the potential therapeutic effect of vitamin D supplementation on the progression of AD pathology will be necessary.
Collapse
Affiliation(s)
- Junha Park
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Korea
| | - Min Soo Byun
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Korea
- Convergence Research Center for Dementia, Seoul National University Medical Research Center, Seoul, Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Seoul National University Medical Research Center, Seoul, Korea
| | - Hyejin Ahn
- Interdisciplinary Program of Cognitive Science, College of Humanities, Seoul National University, Seoul, Korea
| | - Joon Hyung Jung
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea
- Department of Psychiatry, Chungbuk National University Hospital, Cheongju, Korea
| | - Nayeong Kong
- Department of Psychiatry, Keimyung University Dongsan Medical Center, Daegu, Korea
| | - Yoon Young Chang
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea
- Department of Psychiatry, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, Korea
| | - Gijung Jung
- Institute of Human Behavioral Medicine, Seoul National University Medical Research Center, Seoul, Korea
| | - Jun-Young Lee
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea
- Department of Neuropsychiatry, SMG-SNU Boramae Medical Center, Seoul, Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, Korea
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital and Seoul National University College of Medicine, Seoul, Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital and Seoul National University College of Medicine, Seoul, Korea
| | - Dong Young Lee
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Korea
- Convergence Research Center for Dementia, Seoul National University Medical Research Center, Seoul, Korea
- Institute of Human Behavioral Medicine, Seoul National University Medical Research Center, Seoul, Korea
- Interdisciplinary Program of Cognitive Science, College of Humanities, Seoul National University, Seoul, Korea
| | | |
Collapse
|
20
|
Jang H, Chun MY, Yun J, Kim JP, Kang SH, Weiner M, Kim HJ, Na DL, Hong C, Son SJ, Roh HW, Lee T, Lee E, Lee EH, Shin D, Ham H, Gu Y, Kim Y, Kim C, Woo S, Seo SW. Ethnic differences in the prevalence of amyloid positivity and cognitive trajectories. Alzheimers Dement 2024; 20:7556-7566. [PMID: 39315862 PMCID: PMC11567875 DOI: 10.1002/alz.14247] [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: 04/27/2024] [Revised: 07/03/2024] [Accepted: 08/02/2024] [Indexed: 09/25/2024]
Abstract
INTRODUCTION We investigated the prevalence of amyloid beta (Aβ) positivity (+) and cognitive trajectories in Koreans and non-Hispanic Whites (NHWs). METHODS We included 5121 Koreans from multiple centers across South Korea and 929 NHWs from the Alzheimer's Disease Neuroimaging Initiative (ADNI). Participants underwent Aβ positron emission tomography and were categorized into cognitively unimpaired (CU), mild cognitive impairment (MCI), and dementia stages. Age, sex, education, and apolipoprotein E. genotype were adjusted using multivariable logistic regression and stabilized inverse probability of treatment weights based on the propensity scores to mitigate imbalances in these variables. RESULTS The prevalence of Aβ+ was lower in CU Koreans than in CU NHWs (adjusted odds ratio 0.60). Aβ+ Koreans showed a faster cognitive decline than Aβ+ NHWs in the CU (B = -0.314, p = .004) and MCI stages (B = -0.385, p < .001). DISCUSSION Ethnic characteristics of Aβ biomarkers should be considered in research and clinical application of Aβ-targeted therapies in diverse populations. HIGHLIGHTS Koreans have a lower prevalence of Aβ positivity compared to NHWs in the CU stage. The effects of Alzheimer's risk factors on Aβ positivity differ between Koreans and NHWs. Aβ-positive (Aβ+) Koreans show faster cognitive decline than Aβ+ NHWs in the CU and MCI stages.
Collapse
Affiliation(s)
- Hyemin Jang
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineGangnam‐guSeoulSouth Korea
- Department of NeurologySeoul National University HospitalSeoul National University College of MedicineJongno‐guSeoulSouth Korea
| | - Min Young Chun
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineGangnam‐guSeoulSouth Korea
- Department of NeurologyYonsei University College of MedicineSeodaemun‐guSeoulSouth Korea
- Department of NeurologyYongin Severance HospitalYonsei University Health SystemYongin‐siGyeonggi‐doSouth Korea
| | - Jihwan Yun
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineGangnam‐guSeoulSouth Korea
- Department of NeurologySoonchunhyang University Bucheon HospitalBucheon‐siGyeonggi‐doSouth Korea
| | - Jun Pyo Kim
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineGangnam‐guSeoulSouth Korea
- Alzheimer's Disease Convergence Research CenterSamsung Medical CenterGangnam‐guSeoulSouth Korea
| | - Sung Hoon Kang
- Department of NeurologyKorea University Guro HospitalKorea University College of MedicineGuro‐guSeoulSouth Korea
| | - Michael Weiner
- Department of Radiology and Biomedical ImagingUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Hee Jin Kim
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineGangnam‐guSeoulSouth Korea
- Alzheimer's Disease Convergence Research CenterSamsung Medical CenterGangnam‐guSeoulSouth Korea
- Department of Health Sciences and TechnologySAIHST, Sungkyunkwan UniversitySeoulSouth Korea
- Department of Digital HealthSAIHST, Sungkyunkwan UniversityGangnam‐guSeoulSouth Korea
| | - Duk L. Na
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineGangnam‐guSeoulSouth Korea
- Alzheimer's Disease Convergence Research CenterSamsung Medical CenterGangnam‐guSeoulSouth Korea
| | - Chang‐Hyung Hong
- Department of PsychiatryAjou University School of MedicineAjou University HospitalSuwon‐siGyeonggi‐doSouth Korea
| | - Sang Joon Son
- Department of PsychiatryAjou University School of MedicineAjou University HospitalSuwon‐siGyeonggi‐doSouth Korea
| | - Hyun Woong Roh
- Department of PsychiatryAjou University School of MedicineAjou University HospitalSuwon‐siGyeonggi‐doSouth Korea
| | - Tae‐Kyeong Lee
- Department of NeurologySoonchunhyang University Bucheon HospitalBucheon‐siGyeonggi‐doSouth Korea
| | - Eek‐Sung Lee
- Department of NeurologySoonchunhyang University Bucheon HospitalBucheon‐siGyeonggi‐doSouth Korea
| | - Eun Hye Lee
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineGangnam‐guSeoulSouth Korea
| | - Daeun Shin
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineGangnam‐guSeoulSouth Korea
| | - Hongki Ham
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineGangnam‐guSeoulSouth Korea
- Alzheimer's Disease Convergence Research CenterSamsung Medical CenterGangnam‐guSeoulSouth Korea
| | - Yuna Gu
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineGangnam‐guSeoulSouth Korea
- Department of Health Sciences and TechnologySAIHST, Sungkyunkwan UniversitySeoulSouth Korea
| | - Yeshin Kim
- Department of NeurologyKangwon National University School of MedicineGangwon‐doSouth Korea
| | - Chi‐Hun Kim
- Department of NeurologyHallym University Sacred Heart HospitalAnyang‐siGyeonggi‐doSouth Korea
| | - Sook‐young Woo
- Biomedical Statistics CenterData Science Research InstituteSamsung Medical CenterGangnam‐guSeoulSouth Korea
| | - Sang Won Seo
- Department of NeurologySamsung Medical CenterSungkyunkwan University School of MedicineGangnam‐guSeoulSouth Korea
- Alzheimer's Disease Convergence Research CenterSamsung Medical CenterGangnam‐guSeoulSouth Korea
- Department of Health Sciences and TechnologySAIHST, Sungkyunkwan UniversitySeoulSouth Korea
- Department of Digital HealthSAIHST, Sungkyunkwan UniversityGangnam‐guSeoulSouth Korea
| | | |
Collapse
|
21
|
Lin PBC, Holtzman DM. Current insights into apolipoprotein E and the immune response in Alzheimer's disease. Immunol Rev 2024; 327:43-52. [PMID: 39445515 PMCID: PMC11578782 DOI: 10.1111/imr.13414] [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: 10/25/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurological disorder and the most common cause of dementia. Genetic analyses identified apolipoprotein E (APOE) as the strongest genetic risk for late-onset AD. Studies have shown that ApoE modulates AD pathogenesis in part by influencing amyloid-β (Aβ) deposition. However, ApoE also appears to regulate elements of AD via regulation of innate immune response, especially through microglial and astrocyte activation. In model systems, it also regulates changes in T-cells. This review focuses on the key findings that have advanced our understanding of the role of ApoE in the pathogenesis of AD and the current view of innate immune response regulated by ApoE in AD, while discussing open questions and areas for future research.
Collapse
Affiliation(s)
- Peter Bor-Chian Lin
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University, St. Louis, Missouri, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University, St. Louis, Missouri, USA
| |
Collapse
|
22
|
Salardini E, O'Dell RS, Tchorz E, Nabulsi NB, Huang Y, Carson RE, van Dyck CH, Mecca AP. Assessment of the relationship between synaptic density and metabotropic glutamate receptors in early Alzheimer's disease: a multi-tracer PET study. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.21.614277. [PMID: 39386453 PMCID: PMC11463661 DOI: 10.1101/2024.09.21.614277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Background The pathological effects of amyloid β oligomers (Aβo) may be mediated through the metabotropic glutamate receptor subtype 5 (mGluR5), leading to synaptic loss in Alzheimer's disease (AD). Positron emission tomography (PET) studies of mGluR5 using [18F]FPEB indicate a reduction of receptor binding that is focused in the medial temporal lobe in AD. Synaptic loss due to AD measured through synaptic vesicle glycoprotein 2A (SV2A) quantification with [11C]UCB-J PET is also focused in the medial temporal lobe, but with clear widespread reductions is commonly AD-affected neocortical regions. In this study, we used [18F]FPEB and [11C]UCB-J PET to investigate the relationship between mGluR5 and synaptic density in early AD. Methods Fifteen amyloid positive participants with early AD and 12 amyloid negative, cognitively normal (CN) participants underwent PET scans with both [18F]FPEB to measure mGluR5 and [11C]UCB-J to measure synaptic density. Parametric DVR images using equilibrium methods were generated from dynamic. For [18F]FPEB PET, DVR was calculated using equilibrium methods and a cerebellum reference region. For [11C]UCB-J PET, DVR was calculated with a simplified reference tissue model - 2 and a whole cerebellum reference region.. Result A strong positive correlation between mGluR5 and synaptic density was present in the hippocampus for participants with AD (r = 0.81, p < 0.001) and in the CN group (r = 0.74, p = 0.005). In the entorhinal cortex, there was a strong positive correlation between mGluR5 and synaptic in the AD group (r = 0.85, p <0.001), but a weaker non-significant correlation in the CN group (r = 0.36, p = 0.245). Exploratory analyses within and between other brain regions suggested significant positive correlations between mGluR5 in the medial temporal lobe and synaptic density in a broader set of commonly AD-affected regions. Conclusion Medial temporal loss of mGluR5 in AD is associated with synaptic loss in both medial temporal regions and more broadly in association cortical regions, indicating that mGluR5 mediated Aβo toxicity may lead to early synaptic loss more broadly in AD-affected networks. In CN individuals, an isolated strong association between lower mGluR5 and lower synaptic density may indicate non-AD related synaptic loss.
Collapse
Affiliation(s)
- Elaheh Salardini
- Alzheimer's Disease Research Unit, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Ryan S O'Dell
- Alzheimer's Disease Research Unit, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Em Tchorz
- Alzheimer's Disease Research Unit, Yale University School of Medicine, New Haven, CT, USA
| | - Nabeel B Nabulsi
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Christopher H van Dyck
- Alzheimer's Disease Research Unit, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| | - Adam P Mecca
- Alzheimer's Disease Research Unit, Yale University School of Medicine, New Haven, CT, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
23
|
Khajouei E, Ghisays V, Piras IS, Martinez KL, Naymik M, Ngo P, Tran TC, Denny JC, Wheeler TJ, Huentelman MJ, Reiman EM, Karnes JH. Phenome-Wide Association of APOE Alleles in the All of Us Research Program. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.04.24313010. [PMID: 39281754 PMCID: PMC11398429 DOI: 10.1101/2024.09.04.24313010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Background Genetic variation in APOE is associated with altered lipid metabolism, as well as cardiovascular and neurodegenerative disease risk. However, prior studies are largely limited to European ancestry populations and differential risk by sex and ancestry has not been widely evaluated. We utilized a phenome-wide association study (PheWAS) approach to explore APOE-associated phenotypes in the All of Us Research Program. Methods We determined APOE alleles for 181,880 All of Us participants with whole genome sequencing and electronic health record (EHR) data, representing seven gnomAD ancestry groups. We tested association of APOE variants, ordered based on Alzheimer's disease risk hierarchy (ε2/ε2<ε2/ε3<ε3/ε3<ε2/ε4<ε3/ε4<ε4/ε4), with 2,318 EHR-derived phenotypes. Bonferroni-adjusted analyses were performed overall, by ancestry, by sex, and with adjustment for social determinants of health (SDOH). Findings In the overall cohort, PheWAS identified 17 significant associations, including an increased odds of hyperlipidemia (OR 1.15 [1.14-1.16] per APOE genotype group; P=1.8×10-129), dementia, and Alzheimer's disease (OR 1.55 [1.40-1.70]; P=5×10-19), and a reduced odds of fatty liver disease (OR 0.93 [0.90-0.95]; P=1.6×10-9) and chronic liver disease. ORs were similar after SDOH adjustment and by sex, except for an increased number of cardiovascular associations in males, and decreased odds of noninflammatory disorders of vulva and perineum in females (OR 0.89 [0.84-0.94]; P=1.1×10-5). Significant heterogeneity was observed for hyperlipidemia and mild cognitive impairment across ancestry. Unique associations by ancestry included transient retinal arterial occlusion in the European ancestry group, and first-degree atrioventricular block in the American Admixed/Latino ancestry group. Interpretation We replicate extensive phenotypic associations with APOE alleles in a large, diverse cohort, despite limitations in accuracy for EHR-derived phenotypes. We provide a comprehensive catalog of APOE-associated phenotypes and present evidence of unique phenotypic associations by sex and ancestry, as well as heterogeneity in effect size across ancestry.
Collapse
Affiliation(s)
- Ehsan Khajouei
- Department of Pharmacy Practice and Science, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | | | - Ignazio S. Piras
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Kiana L. Martinez
- Department of Pharmacy Practice and Science, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Marcus Naymik
- Neurogenomics Division, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Preston Ngo
- Department of Pharmacy Practice and Science, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Tam C. Tran
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Joshua C. Denny
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
- All of Us Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Travis J. Wheeler
- Department of Pharmacy Practice and Science, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | | | | | - Jason H. Karnes
- Department of Pharmacy Practice and Science, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
24
|
Barnes CA, Permenter MR, Vogt JA, Chen K, Beach TG. Human Alzheimer's Disease ATN/ABC Staging Applied to Aging Rhesus Macaque Brains: Association With Cognition and MRI-Based Regional Gray Matter Volume. J Comp Neurol 2024; 532:e25670. [PMID: 39315417 PMCID: PMC11451939 DOI: 10.1002/cne.25670] [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: 05/29/2024] [Revised: 08/21/2024] [Accepted: 08/30/2024] [Indexed: 09/25/2024]
Abstract
The brain changes of Alzheimer's disease (AD) include Abeta (Aβ) amyloid plaques ("A"), abnormally phosphorylated tau tangles ("T"), and neurodegeneration ("N"). These have been used to construct in vivo and postmortem diagnostic and staging classifications for evaluating the spectrum of AD in the "ATN" and "ABC" ("B" for Braak tau stage, "C" for Consortium to Establish a Registry for Alzheimer's Disease [CERAD] neuritic plaque density) systems. Another common AD feature involves cerebral amyloid angiopathy (CAA). We report the first experiment to examine relationships among cognition, brain distribution of amyloid plaques, CAA, tau/tangles, and magnetic resonance imaging (MRI)-determined volume changes (as a measure of "N") in the same group of behaviorally characterized nonhuman primates. Both ATN and ABC systems were applied to a group of 32 rhesus macaques aged between 7 and 33 years. When an immunohistochemical method for "T" and "B" was used, some monkeys were "triple positive" on ATN, with a maximum ABC status of A1B2C3. With silver or thioflavin S methods, however, all monkeys were classified as T-negative and B0, indicating the absence of mature neurofibrillary tangles (NFTs) and hence neuropathologically defined AD. Although monkeys at extremes of the ATN and ABC classifications, or with frequent CAA, had significantly lower scores on some cognitive tests, the lack of fully mature NFTs or dementia-consistent cognitive impairment indicates that fully developed AD may not occur in rhesus macaques. There were sex differences noted in the types of histopathology present, and only CAA was significantly related to gray matter volume.
Collapse
Affiliation(s)
- Carol A Barnes
- Departments of Psychology, Neurology and Neuroscience, Evelyn F. McKnight Brain Institute, University of Arizona, Tucson, Tucson, Arizona, USA
| | - Michele R Permenter
- California National Primate Center, University of California, Davis, Davis, California, USA
| | - Julie A Vogt
- California National Primate Center, University of California, Davis, Davis, California, USA
| | - Kewei Chen
- Arizona State University, Tempe, Arizona, USA
| | - Thomas G Beach
- Department of Neuroscience, Banner Sun Health Research Institute, Sun City, Arizona, USA
| |
Collapse
|
25
|
Jin S, Wang J, He Y. The brain network hub degeneration in Alzheimer's disease. BIOPHYSICS REPORTS 2024; 10:213-229. [PMID: 39281195 PMCID: PMC11399886 DOI: 10.52601/bpr.2024.230025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 04/26/2024] [Indexed: 09/18/2024] Open
Abstract
Alzheimer's disease (AD) has been conceptualized as a syndrome of brain network dysfunction. Recent imaging connectomics studies have provided unprecedented opportunities to map structural and functional brain networks in AD. By reviewing molecular, imaging, and computational modeling studies, we have shown that highly connected brain hubs are primarily distributed in the medial and lateral prefrontal, parietal, and temporal regions in healthy individuals and that the hubs are selectively and severely affected in AD as manifested by increased amyloid-beta deposition and regional atrophy, hypo-metabolism, and connectivity dysfunction. Furthermore, AD-related hub degeneration depends on the imaging modality with the most notable degeneration in the medial temporal hubs for morphological covariance networks, the prefrontal hubs for structural white matter networks, and in the medial parietal hubs for functional networks. Finally, the AD-related hub degeneration shows metabolic, molecular, and genetic correlates. Collectively, we conclude that the brain-network-hub-degeneration framework is promising to elucidate the biological mechanisms of network dysfunction in AD, which provides valuable information on potential diagnostic biomarkers and promising therapeutic targets for the disease.
Collapse
Affiliation(s)
- Suhui Jin
- Institute for Brain Research and Rehabilitation, Guangzhou 510631, China
| | - Jinhui Wang
- Institute for Brain Research and Rehabilitation, Guangzhou 510631, China
- Guangdong Key Laboratory of Mental Health and Cognitive Science, Guangzhou 510631, China
- Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China
| | - Yong He
- IDG/McGovern Institute for Brain Research, Beijing 100875, China
- National Key Laboratory of Cognitive Neuroscience and Learning, Beijing 100875, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
26
|
Moon HJ, Luo Y, Chugh D, Zhao L. Human apolipoprotein E glycosylation and sialylation: from structure to function. Front Mol Neurosci 2024; 17:1399965. [PMID: 39169951 PMCID: PMC11335735 DOI: 10.3389/fnmol.2024.1399965] [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: 03/12/2024] [Accepted: 06/28/2024] [Indexed: 08/23/2024] Open
Abstract
Human apolipoprotein E (ApoE) was first identified as a polymorphic gene in the 1970s; however, the genetic association of ApoE genotypes with late-onset sporadic Alzheimer's disease (sAD) was only discovered 20 years later. Since then, intensive research has been undertaken to understand the molecular effects of ApoE in the development of sAD. Despite three decades' worth of effort and over 10,000 papers published, the greatest mystery in the ApoE field remains: human ApoE isoforms differ by only one or two amino acid residues; what is responsible for their significantly distinct roles in the etiology of sAD, with ApoE4 conferring the greatest genetic risk for sAD whereas ApoE2 providing exceptional neuroprotection against sAD. Emerging research starts to point to a novel and compelling hypothesis that the sialoglycans posttranslationally appended to human ApoE may serve as a critical structural modifier that alters the biology of ApoE, leading to the opposing impacts of ApoE isoforms on sAD and likely in the peripheral systems as well. ApoE has been shown to be posttranslationally glycosylated in a species-, tissue-, and cell-specific manner. Human ApoE, particularly in brain tissue and cerebrospinal fluid (CSF), is highly glycosylated, and the glycan chains are exclusively attached via an O-linkage to serine or threonine residues. Moreover, studies have indicated that human ApoE glycans undergo sialic acid modification or sialylation, a structural alteration found to be more prominent in ApoE derived from the brain and CSF than plasma. However, whether the sialylation modification of human ApoE has a biological role is largely unexplored. Our group recently first reported that the three major isoforms of human ApoE in the brain undergo varying degrees of sialylation, with ApoE2 exhibiting the most abundant sialic acid modification, whereas ApoE4 is the least sialylated. Our findings further indicate that the sialic acid moiety on human ApoE glycans may serve as a critical modulator of the interaction of ApoE with amyloid β (Aβ) and downstream Aβ pathogenesis, a prominent pathologic feature in AD. In this review, we seek to provide a comprehensive summary of this exciting and rapidly evolving area of ApoE research, including the current state of knowledge and opportunities for future exploration.
Collapse
Affiliation(s)
- Hee-Jung Moon
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Yan Luo
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Diksha Chugh
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Liqin Zhao
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
- Neuroscience Graduate Program, University of Kansas, Lawrence, KS, United States
| |
Collapse
|
27
|
Jackson RJ, Hyman BT, Serrano-Pozo A. Multifaceted roles of APOE in Alzheimer disease. Nat Rev Neurol 2024; 20:457-474. [PMID: 38906999 DOI: 10.1038/s41582-024-00988-2] [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] [Accepted: 05/24/2024] [Indexed: 06/23/2024]
Abstract
For the past three decades, apolipoprotein E (APOE) has been known as the single greatest genetic modulator of sporadic Alzheimer disease (AD) risk, influencing both the average age of onset and the lifetime risk of developing AD. The APOEε4 allele significantly increases AD risk, whereas the ε2 allele is protective relative to the most common ε3 allele. However, large differences in effect size exist across ethnoracial groups that are likely to depend on both global genetic ancestry and local genetic ancestry, as well as gene-environment interactions. Although early studies linked APOE to amyloid-β - one of the two culprit aggregation-prone proteins that define AD - in the past decade, mounting work has associated APOE with other neurodegenerative proteinopathies and broader ageing-related brain changes, such as neuroinflammation, energy metabolism failure, loss of myelin integrity and increased blood-brain barrier permeability, with potential implications for longevity and resilience to pathological protein aggregates. Novel mouse models and other technological advances have also enabled a number of therapeutic approaches aimed at either attenuating the APOEε4-linked increased AD risk or enhancing the APOEε2-linked AD protection. This Review summarizes this progress and highlights areas for future research towards the development of APOE-directed therapeutics.
Collapse
Affiliation(s)
- Rosemary J Jackson
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA.
| | - Alberto Serrano-Pozo
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA.
| |
Collapse
|
28
|
Daly J, De Luca F, Berens SC, Field AP, Rusted JM, Bird CM. The effect of apolipoprotein E genotype on spatial processing in humans: A meta-analysis and systematic review. Cortex 2024; 177:268-284. [PMID: 38878339 DOI: 10.1016/j.cortex.2024.05.006] [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: 01/03/2024] [Revised: 04/04/2024] [Accepted: 05/20/2024] [Indexed: 07/31/2024]
Abstract
The ε4 allele of the apolipoprotein E (APOE4) gene is an established risk factor for Alzheimer's disease but its impact on cognition in healthy adults across the lifespan is unclear. One cognitive domain that is affected early in the course of Alzheimer's disease is spatial cognition, yet the evidence for APOE-related changes in spatial cognition is mixed. In this meta-analysis we assessed the impact of carrying the APOE4 allele on five subdomains of spatial cognition across the lifespan. We included studies of healthy human participants where an APOE4-carrier group (heterozygous or homozygous) could be compared to a homozygous group of APOE3-carriers. We identified 156 studies in total from three databases (Pubmed, Scopus and Web of Science) as well as through searching cited literature and contacting authors for unpublished data. 122 studies involving 32,547 participants were included in a meta-analysis, and the remaining studies are included in a descriptive review. APOE4 carriers scored significantly lower than APOE3 carriers (θˆ = -.08 [-.14, -.02]) on tests of spatial long-term memory; this effect was very small and was not modulated by age. On other subdomains of spatial cognition (spatial construction, spatial working memory, spatial reasoning, navigation) there were no effects of genotype. Overall, our results demonstrate that the APOE4 allele exerts little influence on spatial cognitive abilities in healthy adults.
Collapse
Affiliation(s)
- Jessica Daly
- Sussex Neuroscience, School of Psychology, University of Sussex, United Kingdom
| | - Flavia De Luca
- Sussex Neuroscience, School of Psychology, University of Sussex, United Kingdom
| | - Sam C Berens
- Sussex Neuroscience, School of Psychology, University of Sussex, United Kingdom
| | - Andy P Field
- School of Psychology, University of Sussex, United Kingdom
| | | | - Chris M Bird
- Sussex Neuroscience, School of Psychology, University of Sussex, United Kingdom.
| |
Collapse
|
29
|
Byeon JH, Byun MS, Yi D, Jung JH, Sohn BK, Chang YY, Kong N, Jung G, Ahn H, Lee JY, Lee YS, Kim YK, Lee DY. Moderation of thyroid hormones for the relationship between amyloid and tau pathology. Alzheimers Res Ther 2024; 16:164. [PMID: 39044293 PMCID: PMC11264392 DOI: 10.1186/s13195-024-01534-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 07/16/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND Altered thyroid hormone levels have been associated with increased risk of Alzheimer's disease (AD) dementia and related cognitive decline. However, the neuropathological substrates underlying the link between thyroid hormones and AD dementia are not yet fully understood. We first investigated the association between serum thyroid hormone levels and in vivo AD pathologies including both beta-amyloid (Aβ) and tau deposition measured by positron emission tomography (PET). Given the well-known relationship between Aβ and tau pathology in AD, we additionally examined the moderating effects of thyroid hormone levels on the association between Aβ and tau deposition. METHODS This cross-sectional study was conducted as part of the Korean Brain Aging Study for Early Diagnosis and Prediction of Alzheimer's Disease (KBASE) cohort. This study included a total of 291 cognitively normal adults aged 55 to 90. All participants received comprehensive clinical assessments, measurements for serum total triiodothyronine (T3), free triiodothyronine (fT3), free thyroxine (fT4), and thyroid-stimulating hormone (TSH), and brain imaging evaluations including [11C]-Pittsburgh compound B (PiB)- PET and [18F] AV-1451 PET. RESULTS No associations were found between either thyroid hormones or TSH and Aβ and tau deposition on PET. However, fT4 (p = 0.002) and fT3 (p = 0.001) exhibited significant interactions with Aβ on tau deposition: The sensitivity analyses conducted after the removal of an outlier showed that the interaction effect between fT4 and Aβ deposition was not significant, whereas the interaction between fT3 and Aβ deposition remained significant. However, further subgroup analyses demonstrated a more pronounced positive relationship between Aβ and tau in both the higher fT4 and fT3 groups compared to the lower group, irrespective of outlier removal. Meanwhile, neither T3 nor TSH had any interaction with Aβ on tau deposition. CONCLUSION Our findings suggest that serum thyroid hormones may moderate the relationship between cerebral Aβ and tau pathology. Higher levels of serum thyroid hormones could potentially accelerate the Aβ-dependent tau deposition in the brain. Further replication studies in independent samples are needed to verify the current results.
Collapse
Affiliation(s)
- Jeong Hyeon Byeon
- Department of Neuropsychiatry, Seoul National University Hospital, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Republic of Korea
| | - Min Soo Byun
- Department of Neuropsychiatry, Seoul National University Hospital, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Republic of Korea
- Department of Psychiatry, Seoul National University College of Medicine, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Republic of Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Joon Hyung Jung
- Department of Psychiatry, Chungbuk National University Hospital, Cheongju, Republic of Korea
| | - Bo Kyung Sohn
- Department of Psychiatry, Inje University Sanggye Paik Hospital, Seoul, Republic of Korea
| | - Yoon Young Chang
- Department of Psychiatry, Inje University Sanggye Paik Hospital, Seoul, Republic of Korea
| | - Nayeong Kong
- Department of Psychiatry, Keimyung University Hospital, Daegu, Republic of Korea
| | - Gijung Jung
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Hyejin Ahn
- Interdisciplinary Program of Cognitive Science, Seoul National University, Seoul, Republic of Korea
| | - Jun-Young Lee
- Department of Psychiatry, Seoul National University College of Medicine, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Republic of Korea
- Department of Neuropsychiatry, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea
| | - Dong Young Lee
- Department of Neuropsychiatry, Seoul National University Hospital, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Republic of Korea.
- Department of Psychiatry, Seoul National University College of Medicine, 101 Daehak-Ro, Jongno-Gu, Seoul, 03080, Republic of Korea.
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea.
- Interdisciplinary Program of Cognitive Science, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
30
|
Jones TB, Chu P, Wilkey B, Lynch L, Jentarra G. Regional Differences in Microbial Infiltration of Brain Tissue from Alzheimer's Disease Patients and Control Individuals. Brain Sci 2024; 14:677. [PMID: 39061418 PMCID: PMC11274863 DOI: 10.3390/brainsci14070677] [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: 06/12/2024] [Revised: 06/29/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
Alzheimer's disease (AD) is characterized by cognitive decline and neuropathology including amyloid beta (Aβ) plaques and neurofibrillary tangles (tau). Factors initiating or driving these pathologies remain unclear, though microbes have been increasingly implicated. Our data and others' findings indicate that microbes may be common constituents of the brain. It is notable that Aβ and tau have antimicrobial properties, suggesting a response to microbes in the brain. We used 16S rRNA sequencing to compare major bacterial phyla in post-mortem tissues from individuals exhibiting a range of neuropathology and cognitive status in two brain regions variably affected in AD. Our data indicate that strong regional differences exist, driven in part by the varied presence of Proteobacteria and Firmicutes. We confirmed our data using ELISA of bacterial lipopolysaccharide (LPS) and lipoteichoic acid in the same brain tissue. We identified a potential association between the composition of phyla and the presence of neuropathology but not cognitive status. Declining cognition and increasing pathology correlated closely with serum LPS, but not brain levels of LPS, although brain LPS showed a strong negative correlation with cerebral amyloid angiopathy. Collectively, our data suggest a region-specific heterogeneity of microbial populations in brain tissue potentially associated with neurodegenerative pathology.
Collapse
Affiliation(s)
- T. Bucky Jones
- College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA; (T.B.J.); (P.C.); (L.L.)
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA;
| | - Ping Chu
- College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA; (T.B.J.); (P.C.); (L.L.)
| | - Brooke Wilkey
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA;
- School of Medicine, Creighton University, Phoenix, AZ 85012, USA
| | - Leigha Lynch
- College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA; (T.B.J.); (P.C.); (L.L.)
| | - Garilyn Jentarra
- College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA; (T.B.J.); (P.C.); (L.L.)
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA;
| |
Collapse
|
31
|
Li J, Huang Q, Qi N, He K, Li S, Huang L, Pan F, Ren S, Hua F, Huang Y, Guan Y, Guo Q, Zhao J, Xie F. The associations between synaptic density and "A/T/N" biomarkers in Alzheimer's disease: An 18F-SynVesT-1 PET/MR study. J Cereb Blood Flow Metab 2024; 44:1199-1207. [PMID: 38295871 PMCID: PMC11179616 DOI: 10.1177/0271678x241230733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 12/08/2023] [Accepted: 12/26/2023] [Indexed: 06/13/2024]
Abstract
A newly developed SV2A radiotracer, 18F-SynVesT-1, was used in this study to investigate synaptic density and its association with Alzheimer's disease (AD) "A/T/N" biomarkers. The study included a cohort of 97 subjects, consisting of 64 patients with cognitive impairment (CI) and 33 individuals with normal cognition (CU). All subjects underwent 18F-SynVesT-1 PET/MR and 18F-florbetapir PET/CT scans. Additionally, a subgroup of individuals also underwent 18F-MK-6240, 18F-FDG PET/CT, plasma Aβ42/Aβ40 and p-tau181 tests. The differences in synaptic density between the groups and the correlations between synaptic density and AD "A/T/N" biomarkers were analyzed. The results showed that compared to the CU group, the CI with Aβ+ (CI+) group exhibited the most pronounced synapse loss in the hippocampus, with some loss also observed in the neocortex. Furthermore, synaptic density in the hippocampus and parahippocampal gyrus showed associations with AD biomarkers detected by both imaging and plasma tests in the CI group. The associations between synaptic density and FDG uptake and hippocampal volume were also observed in the CI+ group. In conclusion, the study demonstrated significant synaptic density loss, as measured by the promising tracer 18F-SynVesT-1, and its close correlation with "A/T/N" biomarkers in patients with both Alzheimer's clinical syndrome and pathological changes.
Collapse
Affiliation(s)
- Junpeng Li
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qi Huang
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Na Qi
- Department of Nuclear Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Kun He
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Songye Li
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Lin Huang
- Department of Gerontology, Shanghai Jiaotong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Fengfeng Pan
- Department of Gerontology, Shanghai Jiaotong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Shuhua Ren
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Fengchun Hua
- Department of Nuclear Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiyun Huang
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Yihui Guan
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Qihao Guo
- Department of Gerontology, Shanghai Jiaotong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Jun Zhao
- Department of Nuclear Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
32
|
Ahmed H, Wang Y, Griffiths WJ, Levey AI, Pikuleva I, Liang SH, Haider A. Brain cholesterol and Alzheimer's disease: challenges and opportunities in probe and drug development. Brain 2024; 147:1622-1635. [PMID: 38301270 PMCID: PMC11068113 DOI: 10.1093/brain/awae028] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 12/20/2023] [Accepted: 01/13/2024] [Indexed: 02/03/2024] Open
Abstract
Cholesterol homeostasis is impaired in Alzheimer's disease; however, attempts to modulate brain cholesterol biology have not translated into tangible clinical benefits for patients to date. Several recent milestone developments have substantially improved our understanding of how excess neuronal cholesterol contributes to the pathophysiology of Alzheimer's disease. Indeed, neuronal cholesterol was linked to the formation of amyloid-β and neurofibrillary tangles through molecular pathways that were recently delineated in mechanistic studies. Furthermore, remarkable advances in translational molecular imaging have now made it possible to probe cholesterol metabolism in the living human brain with PET, which is an important prerequisite for future clinical trials that target the brain cholesterol machinery in Alzheimer's disease patients-with the ultimate aim being to develop disease-modifying treatments. This work summarizes current concepts of how the biosynthesis, transport and clearance of brain cholesterol are affected in Alzheimer's disease. Further, current strategies to reverse these alterations by pharmacotherapy are critically discussed in the wake of emerging translational research tools that support the assessment of brain cholesterol biology not only in animal models but also in patients with Alzheimer's disease.
Collapse
Affiliation(s)
- Hazem Ahmed
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, Yale University, New Haven, CT 06510, USA
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, 8093 Zurich, Switzerland
| | - Yuqin Wang
- Institute of Life Science, Swansea University Medical School, Swansea SA2 8PP, UK
| | - William J Griffiths
- Institute of Life Science, Swansea University Medical School, Swansea SA2 8PP, UK
| | - Allan I Levey
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Irina Pikuleva
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Steven H Liang
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ahmed Haider
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA 30322, USA
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
33
|
Lee S, Byun MS, Yi D, Ahn H, Jung G, Jung JH, Chang YY, Kim K, Choi H, Choi J, Lee JY, Kang KM, Sohn CH, Lee YS, Kim YK, Lee DY. Plasma Leptin and Alzheimer Protein Pathologies Among Older Adults. JAMA Netw Open 2024; 7:e249539. [PMID: 38700863 PMCID: PMC11069086 DOI: 10.1001/jamanetworkopen.2024.9539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 03/01/2024] [Indexed: 05/06/2024] Open
Abstract
Importance Many epidemiologic studies have suggested that low levels of plasma leptin, a major adipokine, are associated with increased risk of Alzheimer disease (AD) dementia and cognitive decline. Nevertheless, the mechanistic pathway linking plasma leptin and AD-related cognitive decline is not yet fully understood. Objective To examine the association of plasma leptin levels with in vivo AD pathologies, including amyloid-beta (Aβ) and tau deposition, through both cross-sectional and longitudinal approaches among cognitively unimpaired older adults. Design, Setting, and Participants This was a longitudinal cohort study from the Korean Brain Aging Study for Early Diagnosis and Prediction of Alzheimer Disease. Data were collected from January 1, 2014, to December 31, 2020, and data were analyzed from July 11 to September 6, 2022. The study included a total of 208 cognitively unimpaired participants who underwent baseline positron emission tomography (PET) scans for brain Aβ deposition. For longitudinal analyses, 192 participants who completed both baseline and 2-year follow-up PET scans for brain Aβ deposition were included. Exposure Plasma leptin levels as assessed by enzyme-linked immunosorbent assay. Main Outcomes and Measures Baseline levels and longitudinal changes of global Aβ and AD-signature region tau deposition measured by PET scans. Results Among the 208 participants, the mean (SD) age was 66.0 (11.3) years, 114 were women (54.8%), and 37 were apolipoprotein E ε4 carriers (17.8%). Lower plasma leptin levels had a significant cross-sectional association with greater brain Aβ deposition (β = -0.04; 95% CI, -0.09 to 0.00; P = .046), while there was no significant association between plasma leptin levels and tau deposition (β = -0.02; 95% CI, -0.05 to 0.02; P = .41). In contrast, longitudinal analyses revealed that there was a significant association between lower baseline leptin levels and greater increase of tau deposition over 2 years (β = -0.06; 95% CI, -0.11 to -0.01; P = .03), whereas plasma leptin levels did not have a significant association with longitudinal change of Aβ deposition (β = 0.006; 95% CI, 0.00-0.02; P = .27). Conclusions and Relevance The present findings suggest that plasma leptin may be protective for the development or progression of AD pathology, including both Aβ and tau deposition.
Collapse
Affiliation(s)
- Seunghoon Lee
- Department of Psychiatry, Myongji Hospital, Hanyang University College of Medicine, Goyang, Republic of Korea
| | - Min Soo Byun
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Hyejin Ahn
- Interdisciplinary Program of Cognitive Science, Seoul National University College of Humanities, Seoul, Republic of Korea
| | - Gijung Jung
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Joon Hyung Jung
- Department of Psychiatry, Chungbuk National University Hospital, Cheongju, Republic of Korea
| | - Yoon Young Chang
- Department of Psychiatry, Inje University, Sanggye Paik Hospital, Seoul, Republic of Korea
| | - Kyungtae Kim
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyeji Choi
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jeongmin Choi
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jun-Young Lee
- Department of Neuropsychiatry, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea
| | - Dong Young Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
- Interdisciplinary Program of Cognitive Science, Seoul National University College of Humanities, Seoul, Republic of Korea
| |
Collapse
|
34
|
Ingannato A, Bagnoli S, Mazzeo S, Giacomucci G, Bessi V, Ferrari C, Sorbi S, Nacmias B. Plasma GFAP, NfL and pTau 181 detect preclinical stages of dementia. Front Endocrinol (Lausanne) 2024; 15:1375302. [PMID: 38654932 PMCID: PMC11035722 DOI: 10.3389/fendo.2024.1375302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
Background Plasma biomarkers are preferable to invasive and expensive diagnostic tools, such as neuroimaging and lumbar puncture that are gold standard in the clinical management of Alzheimer's Disease (AD). Here, we investigated plasma Glial Fibrillary Acidic Protein (GFAP), Neurofilament Light Chain (NfL) and Phosphorylated-tau-181 (pTau 181) in AD and in its early stages: Subjective cognitive decline (SCD) and Mild cognitive impairment (MCI). Material and methods This study included 152 patients (42 SCD, 74 MCI and 36 AD). All patients underwent comprehensive clinical and neurological assessment. Blood samples were collected for Apolipoprotein E (APOE) genotyping and plasma biomarker (GFAP, NfL, and pTau 181) measurements. Forty-three patients (7 SCD, 27 MCI, and 9 AD) underwent a follow-up (FU) visit after 2 years, and a second plasma sample was collected. Plasma biomarker levels were detected using the Simoa SR-X technology (Quanterix Corp.). Statistical analysis was performed using SPSS software version 28 (IBM SPSS Statistics). Statistical significance was set at p < 0.05. Results GFAP, NfL and pTau 181 levels in plasma were lower in SCD and MCI than in AD patients. In particular, plasma GFAP levels were statistically significant different between SCD and AD (p=0.003), and between MCI and AD (p=0.032). Plasma NfL was different in SCD vs MCI (p=0.026), SCD vs AD (p<0.001), SCD vs AD FU (p<0.001), SCD FU vs AD (p=0.033), SCD FU vs AD FU (p=0.011), MCI vs AD (p=0.002), MCI FU vs AD (p=0.003), MCI FU vs AD FU (p=0.003) and MCI vs AD FU (p=0.003). Plasma pTau 181 concentration was significantly different between SCD and AD (p=0.001), MCI and AD (p=0.026), MCI FU and AD (p=0.020). In APOE ϵ4 carriers, a statistically significant increase in plasma NfL (p<0.001) and pTau 181 levels was found (p=0.014). Moreover, an association emerged between age at disease onset and plasma GFAP (p = 0.021) and pTau181 (p < 0.001) levels. Discussion and conclusions Plasma GFAP, NfL and pTau 181 are promising biomarkers in the diagnosis of the prodromic stages and prognosis of dementia.
Collapse
Affiliation(s)
- Assunta Ingannato
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Silvia Bagnoli
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Salvatore Mazzeo
- Research and Innovation Centre for Dementia-CRIDEM, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
- Vita-Salute San Raffaele University, Milan, Italy
- Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Donato, San Donato Milanese, Italy
| | - Giulia Giacomucci
- Research and Innovation Centre for Dementia-CRIDEM, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Valentina Bessi
- Research and Innovation Centre for Dementia-CRIDEM, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Camilla Ferrari
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
- Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
- Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Don Carlo Gnocchi, Florence, Italy
| |
Collapse
|
35
|
Strickland MR, Rau MJ, Summers B, Basore K, Wulf J, Jiang H, Chen Y, Ulrich JD, Randolph GJ, Zhang R, Fitzpatrick JAJ, Cashikar AG, Holtzman DM. Apolipoprotein E secreted by astrocytes forms antiparallel dimers in discoidal lipoproteins. Neuron 2024; 112:1100-1109.e5. [PMID: 38266643 PMCID: PMC10994765 DOI: 10.1016/j.neuron.2023.12.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/24/2023] [Accepted: 12/22/2023] [Indexed: 01/26/2024]
Abstract
The Apolipoprotein E gene (APOE) is of great interest due to its role as a risk factor for late-onset Alzheimer's disease. ApoE is secreted by astrocytes in the central nervous system in high-density lipoprotein (HDL)-like lipoproteins. Structural models of lipidated ApoE of high resolution could aid in a mechanistic understanding of how ApoE functions in health and disease. Using monoclonal Fab and F(ab')2 fragments, we characterize the structure of lipidated ApoE on astrocyte-secreted lipoproteins. Our results provide support for the "double-belt" model of ApoE in nascent discoidal HDL-like lipoproteins, where two ApoE proteins wrap around the nanodisc in an antiparallel conformation. We further show that lipidated, recombinant ApoE accurately models astrocyte-secreted ApoE lipoproteins. Cryogenic electron microscopy of recombinant lipidated ApoE further supports ApoE adopting antiparallel dimers in nascent discoidal lipoproteins.
Collapse
Affiliation(s)
| | - Michael J Rau
- Washington University Center for Cellular Imaging, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Brock Summers
- Washington University Center for Cellular Imaging, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Katherine Basore
- Washington University Center for Cellular Imaging, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - John Wulf
- Washington University Center for Cellular Imaging, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Hong Jiang
- Department of Neurology, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Yun Chen
- Department of Neurology, 660 S. Euclid Ave., St. Louis, MO 63110, USA; Department of Pathology and Immunology, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Jason D Ulrich
- Department of Neurology, 660 S. Euclid Ave., St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, 660 S. Euclid Ave., St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, 4488 Forest Park Ave., St. Louis, MO 63108, USA
| | - Gwendalyn J Randolph
- Department of Pathology and Immunology, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Rui Zhang
- Department of Biochemistry and Molecular Biophysics, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - James A J Fitzpatrick
- Washington University Center for Cellular Imaging, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Anil G Cashikar
- Hope Center for Neurological Disorders, 660 S. Euclid Ave., St. Louis, MO 63110, USA; Department of Psychiatry, 660 S. Euclid Ave., St. Louis, MO 63110, USA; Taylor Family institute for Innovative Psychiatric Research, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - David M Holtzman
- Department of Neurology, 660 S. Euclid Ave., St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, 660 S. Euclid Ave., St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, 4488 Forest Park Ave., St. Louis, MO 63108, USA.
| |
Collapse
|
36
|
Rasmussen KL, Frikke-Schmidt R. The current state of apolipoprotein E in dyslipidemia. Curr Opin Lipidol 2024; 35:78-84. [PMID: 38054895 DOI: 10.1097/mol.0000000000000915] [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: 12/07/2023]
Abstract
PURPOSE OF REVIEW Apolipoprotein E (apoE) plays a pivotal role in lipid metabolism in the peripheral circulation and in the brain. This has been recognized for decades; however, the importance of the full spectrum of variation in the APOE gene has been less investigated. This review focusses on current progresses in this field with main focus on apoE in dyslipidemia and vascular disease. RECENT FINDINGS Whereas ε4 is the risk increasing allele for Alzheimer disease, ε2 is associated with increased risk for age-related macular degeneration. Rare functional ε2-like variants in APOE have previously been reported to have protective associations for Alzheimer disease but recent findings suggest a simultaneous high risk of age-related macular degeneration, in line with observations for the ε2 allele. SUMMARY ApoE plays an important and well established role in dyslipidemia, vascular disease, and dementia. Recent evidence from large general population studies now also suggests that apoE is involved in age-related macular degeneration. ApoE-targeted therapeutics are being developed for multiple purposes; this heralds a promising change in the approach to disease processes involving apoE. The different risk profile for dementia and age-related macular degeneration should, however, be kept in mind when developing drugs targeting mechanisms resembling these variants.
Collapse
Affiliation(s)
- Katrine L Rasmussen
- Department of Clinical Biochemistry, Nordsjællands Hospital, Hillerød
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Herlev
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
| | - Ruth Frikke-Schmidt
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Herlev
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
37
|
Vance JM, Farrer LA, Huang Y, Cruchaga C, Hyman BT, Pericak-Vance MA, Goate AM, Greicius MD, Griswold AJ, Haines JL, Tcw J, Schellenberg GD, Tsai LH, Herz J, Holtzman DM. Report of the APOE4 National Institute on Aging/Alzheimer Disease Sequencing Project Consortium Working Group: Reducing APOE4 in Carriers is a Therapeutic Goal for Alzheimer's Disease. Ann Neurol 2024; 95:625-634. [PMID: 38180638 DOI: 10.1002/ana.26864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 12/06/2023] [Accepted: 12/09/2023] [Indexed: 01/06/2024]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder and one of the leading causes of disability worldwide. The apolipoprotein E4 gene (APOE4) is the strongest genetic risk factor for AD. In 2023, the APOE4 National Institute on Aging/Alzheimer's Disease Sequencing Project working group came together to gather data and discuss the question of whether to reduce or increase APOE4 as a therapeutic intervention for AD. It was the unanimous consensus that cumulative data from multiple studies in humans and animal models support that lowering APOE4 should be a target for therapeutic approaches for APOE4 carriers. ANN NEUROL 2024;95:625-634.
Collapse
Affiliation(s)
- Jeffery M Vance
- John T. McDonald Department of Human Genetics, John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Lindsay A Farrer
- Departments of Medicine (Biomedical Genetics), Neurology and Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Departments of Epidemiology and Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Yadong Huang
- Department of Neurology, Gladstone Center for Translational Advancement, Gladstone Institute of Neurological Disease, University of California, San Francisco, San Francisco, CA, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, USA
| | - Bradley T Hyman
- Alzheimer Research Unit, Department of Neurology, The Massachusetts General Hospital Institute for Neurodegenerative Disease, Harvard Medical School, Boston, MA, USA
| | - Margaret A Pericak-Vance
- John T. McDonald Department of Human Genetics, John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Alison M Goate
- Departments of Genetics & Genomic Sciences, Ronald M. Loeb Center for Alzheimer's disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael D Greicius
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Anthony J Griswold
- John P. Hussman Institute for Human Genomics, The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jonathan L Haines
- Department of Population & Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Julia Tcw
- Departments of Pharmacology, Physiology & Biophysics, Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Bioinformatics Program, Faculty of Computing & Data Sciences, Boston University, Boston, MA, USA
| | - Gerard D Schellenberg
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Li-Huei Tsai
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Joachim Herz
- Departments of Molecular Genetics, Neuroscience, Neurology, Center for Translational Neurodegeneration Research, UT Southwestern, Dallas, TX, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
38
|
Cha WJ, Yi D, Ahn H, Byun MS, Chang YY, Choi JM, Kim K, Choi H, Jung G, Kang KM, Sohn CH, Lee YS, Kim YK, Lee DY. Association between brain amyloid deposition and longitudinal changes of white matter hyperintensities. Alzheimers Res Ther 2024; 16:50. [PMID: 38454444 PMCID: PMC10918927 DOI: 10.1186/s13195-024-01417-8] [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: 11/04/2023] [Accepted: 02/20/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND Growing evidence suggests that not only cerebrovascular disease but also Alzheimer's disease (AD) pathological process itself cause cerebral white matter degeneration, resulting in white matter hyperintensities (WMHs). Some preclinical evidence also indicates that white matter degeneration may precede or affect the development of AD pathology. This study aimed to clarify the direction of influence between in vivo AD pathologies, particularly beta-amyloid (Aβ) and tau deposition, and WMHs through longitudinal approach. METHODS Total 282 older adults including cognitively normal and cognitively impaired individuals were recruited from the Korean Brain Aging Study for the Early Diagnosis and Prediction of Alzheimer's Disease (KBASE) cohort. The participants underwent comprehensive clinical and neuropsychological assessment, [11C] Pittsburgh Compound B PET for measuring Aβ deposition, [18F] AV-1451 PET for measuring tau deposition, and MRI scans with fluid-attenuated inversion recovery image for measuring WMH volume. The relationships between Aβ or tau deposition and WMH volume were examined using multiple linear regression analysis. In this analysis, baseline Aβ or tau were used as independent variables, and change of WMH volume over 2 years was used as dependent variable to examine the effect of AD pathology on increase of WMH volume. Additionally, we set baseline WMH volume as independent variable and longitudinal change of Aβ or tau deposition for 2 years as dependent variables to investigate whether WMH volume could precede AD pathologies. RESULTS Baseline Aβ deposition, but not tau deposition, had significant positive association with longitudinal change of WMH volume over 2 years. Baseline WMH volume was not related with any of longitudinal change of Aβ or tau deposition for 2 years. We also found a significant interaction effect between baseline Aβ deposition and sex on longitudinal change of WMH volume. Subsequent subgroup analyses showed that high baseline Aβ deposition was associated with increase of WMH volume over 2 years in female, but not in male. CONCLUSIONS Our findings suggest that Aβ deposition accelerates cerebral WMHs, particularly in female, whereas white matter degeneration appears not influence on longitudinal Aβ increase. The results also did not support any direction of influence between tau deposition and WMHs.
Collapse
Affiliation(s)
- Woo-Jin Cha
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Hyejin Ahn
- Interdisciplinary program of cognitive science, Seoul National University College of Humanities, Seoul, Republic of Korea
| | - Min Soo Byun
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yoon Young Chang
- Department of Psychiatry, Inje University Sanggye Paik Hospital, Seoul, Republic of Korea
| | - Jung-Min Choi
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kyungtae Kim
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyeji Choi
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Gijung Jung
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Dong Young Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea.
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea.
- Interdisciplinary program of cognitive science, Seoul National University College of Humanities, Seoul, Republic of Korea.
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
39
|
Niu X, Wang Y, Zhang X, Wang Y, Shao W, Chen L, Yang Z, Peng D. Quantitative electroencephalography (qEEG), apolipoprotein A-I (APOA-I), and apolipoprotein epsilon 4 (APOE ɛ4) alleles for the diagnosis of mild cognitive impairment and Alzheimer's disease. Neurol Sci 2024; 45:547-556. [PMID: 37673807 DOI: 10.1007/s10072-023-07028-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 08/19/2023] [Indexed: 09/08/2023]
Abstract
INTRODUCTION Alzheimer's disease (AD) is the most common type of dementia. Amnestic mild cognitive impairment (aMCI), a pre-dementia stage is an important stage for early diagnosis and intervention. This study aimed to investigate the diagnostic value of qEEG, APOA-I, and APOE ɛ4 allele in aMCI and AD patients and found the correlation between qEEG (Delta + Theta)/(Alpha + Beta) ratio (DTABR) and different cognitive domains. METHODS All participants were divided into three groups: normal controls (NCs), aMCI, and AD, and all received quantitative electroencephalography (qEEG), neuropsychological scale assessment, apolipoprotein epsilon 4 (APOE ɛ4) alleles, and various blood lipid indicators. Different statistical methods were used for different data. RESULTS The cognitive domains except executive ability were all negatively correlated with DTABR in different brain regions while executive ability was positively correlated with DTABR in several brain regions, although without statistical significance. The consequences confirmed that the DTABR of each brain area were related to MMSE, MoCA, instantaneous memory, and the language ability (p < 0.05), and the DTABR in the occipital area was relevant to all cognitive domains (p < 0.01) except executive function (p = 0.272). Also, occipital DTABR was most correlated with language domain when tested by VFT with a moderate level (r = 0.596, p < 0.001). There were significant differences in T3, T5, and P3 DTABR between both AD and NC and aMCI and NCs. As for aMCI diagnosis, the maximum AUC was achieved when using T3 combined with APOA-I and APOE ε4 (0.855) and the maximum AUC was achieved when using T5 combined with APOA-I and APOE ε4 (0.889) for AD diagnosis. CONCLUSION These findings highlight that APOA-I, APOE ɛ4, and qEEG play an important role in aMCI and AD diagnosis. During AD continuum, qEEG DTABR should be taken into consideration for the early detection of AD risk.
Collapse
Affiliation(s)
- Xiaoqian Niu
- Department of Neurology, China-Japan Friendship Hospital, No. 2 Yinghuayuan East Street, Chaoyang District, Beijing, 100029, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Yuye Wang
- Department of Neurology, China-Japan Friendship Hospital, No. 2 Yinghuayuan East Street, Chaoyang District, Beijing, 100029, China
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiangfei Zhang
- Department of Neurology, China-Japan Friendship Hospital, No. 2 Yinghuayuan East Street, Chaoyang District, Beijing, 100029, China
| | - Yu Wang
- Department of Neurology, China-Japan Friendship Hospital, No. 2 Yinghuayuan East Street, Chaoyang District, Beijing, 100029, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Wen Shao
- Department of Neurology, China-Japan Friendship Hospital, No. 2 Yinghuayuan East Street, Chaoyang District, Beijing, 100029, China
| | - Leian Chen
- Department of Neurology, China-Japan Friendship Hospital, No. 2 Yinghuayuan East Street, Chaoyang District, Beijing, 100029, China
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ziyuan Yang
- Department of Neurology, China-Japan Friendship Hospital, No. 2 Yinghuayuan East Street, Chaoyang District, Beijing, 100029, China
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Dantao Peng
- Department of Neurology, China-Japan Friendship Hospital, No. 2 Yinghuayuan East Street, Chaoyang District, Beijing, 100029, China.
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China.
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
40
|
Ofori E, Vaillancourt DE, Greig-Custo MT, Barker W, Hanson K, DeKosky ST, Garvan CS, Adjouadi M, Golde T, Loewenstein DA, Stecher C, Fowers R, Duara R. Free-water imaging reveals unique brain microstructural deficits in hispanic individuals with Dementia. Brain Imaging Behav 2024; 18:106-116. [PMID: 37903991 PMCID: PMC11157151 DOI: 10.1007/s11682-023-00819-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] [Accepted: 10/22/2023] [Indexed: 11/01/2023]
Abstract
Prior evidence suggests that Hispanic and non-Hispanic individuals differ in potential risk factors for the development of dementia. Here we determine whether specific brain regions are associated with cognitive performance for either ethnicity along various stages of Alzheimer's disease. For this cross-sectional study, we examined 108 participants (61 Hispanic vs. 47 Non-Hispanic individuals) from the 1Florida Alzheimer's Disease Research Center (1Florida ADRC), who were evaluated at baseline with diffusion-weighted and T1-weighted imaging, and positron emission tomography (PET) amyloid imaging. We used FreeSurfer to segment 34 cortical regions of interest. Baseline Mini-Mental State Examination (MMSE) and Montreal Cognitive Assessment (MoCA) were used as measures of cognitive performance. Group analyses assessed free-water measures (FW) and volume. Statistically significant FW regions based on ethnicity x group interactions were used in a stepwise regression function to predict total MMSE and MoCA scores. Random forest models were used to identify the most predictive brain-based measures of a dementia diagnosis separately for Hispanic and non-Hispanic groups. Results indicated elevated FW values for the left inferior temporal gyrus, left middle temporal gyrus, left banks of the superior temporal sulcus, left supramarginal gyrus, right amygdala, and right entorhinal cortex in Hispanic AD subjects compared to non-Hispanic AD subjects. These alterations occurred in the absence of different volumes of these regions in the two AD groups. FW may be useful in detecting individual differences potentially reflective of varying etiology that can influence cognitive decline and identify MRI predictors of cognitive performance, particularly among Hispanics.
Collapse
Affiliation(s)
- Edward Ofori
- College of Health Solutions, Arizona State University, 425 N. 5th St Phoenix, Phoenix, AZ, 85004, USA.
| | - David E Vaillancourt
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Maria T Greig-Custo
- Wien Center for Alzheimer's Disease and Memory Disorders, Mount Sinai Medical Center, Miami, FL, USA
| | - Warren Barker
- Wien Center for Alzheimer's Disease and Memory Disorders, Mount Sinai Medical Center, Miami, FL, USA
| | - Kevin Hanson
- Clinical and Translational Science Institute, University of Florida, Gainesville, FL, USA
| | - Steven T DeKosky
- Emory Center for Neurodegenerative Disease, Departments of Pharmacology, Chemical Biology, & Neurology, Atlanta, GA, USA
| | - Cynthia S Garvan
- Department of Anesthesiology, University of Florida, Gainesville, FL, USA
| | - Malek Adjouadi
- Electrical and Computer Engineering, Florida International University, Miami, FL, USA
| | - Todd Golde
- Emory Center for Neurodegenerative Disease, Departments of Pharmacology, Chemical Biology, & Neurology, Atlanta, GA, USA
- Department of Psychiatry, Miller School of Medicine, Center for Cognitive Neuroscience and Aging University of Miami, Miami, FL, USA
| | - David A Loewenstein
- Department of Psychiatry, Miller School of Medicine, Center for Cognitive Neuroscience and Aging University of Miami, Miami, FL, USA
| | - Chad Stecher
- College of Health Solutions, Arizona State University, 425 N. 5th St Phoenix, Phoenix, AZ, 85004, USA
| | - Rylan Fowers
- College of Health Solutions, Arizona State University, 425 N. 5th St Phoenix, Phoenix, AZ, 85004, USA
| | - Ranjan Duara
- Wien Center for Alzheimer's Disease and Memory Disorders, Mount Sinai Medical Center, Miami, FL, USA
| |
Collapse
|
41
|
van Dyck CH, Mecca AP, O'Dell RS, Bartlett HH, Diepenbrock NG, Huang Y, Hamby ME, Grundman M, Catalano SM, Caggiano AO, Carson RE. A pilot study to evaluate the effect of CT1812 treatment on synaptic density and other biomarkers in Alzheimer's disease. Alzheimers Res Ther 2024; 16:20. [PMID: 38273408 PMCID: PMC10809445 DOI: 10.1186/s13195-024-01382-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 01/01/2024] [Indexed: 01/27/2024]
Abstract
BACKGROUND Effective, disease-modifying therapeutics for the treatment of Alzheimer's disease (AD) remain a large unmet need. Extensive evidence suggests that amyloid beta (Aβ) is central to AD pathophysiology, and Aβ oligomers are among the most toxic forms of Aβ. CT1812 is a novel brain penetrant sigma-2 receptor ligand that interferes with the binding of Aβ oligomers to neurons. Preclinical studies of CT1812 have demonstrated its ability to displace Aβ oligomers from neurons, restore synapses in cell cultures, and improve cognitive measures in mouse models of AD. CT1812 was found to be generally safe and well tolerated in a placebo-controlled phase 1 clinical trial in healthy volunteers and phase 1a/2 clinical trials in patients with mild to moderate dementia due to AD. The unique objective of this study was to incorporate synaptic positron emission tomography (PET) imaging as an outcome measure for CT1812 in AD patients. METHODS The present phase 1/2 study was a randomized, double-blind, placebo-controlled, parallel-group trial conducted in 23 participants with mild to moderate dementia due to AD to primarily evaluate the safety of CT1812 and secondarily its pharmacodynamic effects. Participants received either placebo or 100 mg or 300 mg per day of oral CT1812 for 24 weeks. Pharmacodynamic effects were assessed using the exploratory efficacy endpoints synaptic vesicle glycoprotein 2A (SV2A) PET, fluorodeoxyglucose (FDG) PET, volumetric MRI, cognitive clinical measures, as well as cerebrospinal fluid (CSF) biomarkers of AD pathology and synaptic degeneration. RESULTS No treatment differences relative to placebo were observed in the change from baseline at 24 weeks in either SV2A or FDG PET signal, the cognitive clinical rating scales, or in CSF biomarkers. Composite region volumetric MRI revealed a trend towards tissue preservation in participants treated with either dose of CT1812, and nominally significant differences with both doses of CT1812 compared to placebo were found in the pericentral, prefrontal, and hippocampal cortices. CT1812 was safe and well tolerated. CONCLUSIONS The safety findings of this 24-week study and the observed changes on volumetric MRI with CT1812 support its further clinical development. TRIAL REGISTRATION The clinical trial described in this manuscript is registered at clinicaltrials.gov (NCT03493282).
Collapse
Affiliation(s)
- Christopher H van Dyck
- Alzheimer's Disease Research Unit, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.
| | - Adam P Mecca
- Alzheimer's Disease Research Unit, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Ryan S O'Dell
- Alzheimer's Disease Research Unit, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Hugh H Bartlett
- Alzheimer's Disease Research Unit, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Nina G Diepenbrock
- Alzheimer's Disease Research Unit, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Mary E Hamby
- Cognition Therapeutics Inc., Pittsburgh, PA, USA
| | - Michael Grundman
- Global R&D Partners, LLC, San Diego, CA, USA
- Department of Neurosciences, University of California, San Diego, USA
| | | | | | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| |
Collapse
|
42
|
Shukla D, Suryavanshi A, Bharti SK, Asati V, Mahapatra DK. Recent Advances in the Treatment and Management of Alzheimer's Disease: A Precision Medicine Perspective. Curr Top Med Chem 2024; 24:1699-1737. [PMID: 38566385 DOI: 10.2174/0115680266299847240328045737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/20/2024] [Accepted: 02/29/2024] [Indexed: 04/04/2024]
Abstract
About 60% to 70% of people with dementia have Alzheimer's Disease (AD), a neurodegenerative illness. One reason for this disorder is the misfolding of naturally occurring proteins in the human brain, specifically β-amyloid (Aβ) and tau. Certain diagnostic imaging techniques, such as amyloid PET imaging, tau PET imaging, Magnetic Resonance Imaging (MRI), Computerized Tomography (CT), and others, can detect biomarkers in blood, plasma, and cerebral spinal fluids, like an increased level of β-amyloid, plaques, and tangles. In order to create new pharmacotherapeutics for Alzheimer's disease, researchers must have a thorough and detailed knowledge of amyloid beta misfolding and other related aspects. Donepezil, rivastigmine, galantamine, and other acetylcholinesterase inhibitors are among the medications now used to treat Alzheimer's disease. Another medication that can temporarily alleviate dementia symptoms is memantine, which blocks the N-methyl-D-aspartate (NMDA) receptor. However, it is not able to halt or reverse the progression of the disease. Medication now on the market can only halt its advancement, not reverse it. Interventions to alleviate behavioral and psychological symptoms, exhibit anti- neuroinflammation and anti-tau effects, induce neurotransmitter alteration and cognitive enhancement, and provide other targets have recently been developed. For some Alzheimer's patients, the FDA-approved monoclonal antibody, aducanumab, is an option; for others, phase 3 clinical studies are underway for drugs, like lecanemab and donanemab, which have demonstrated potential in eliminating amyloid protein. However, additional study is required to identify and address these limitations in order to reduce the likelihood of side effects and maximize the therapeutic efficacy.
Collapse
Affiliation(s)
- Deepali Shukla
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Anjali Suryavanshi
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Sanjay Kumar Bharti
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Vivek Asati
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| | - Debarshi Kar Mahapatra
- Department of Pharmaceutical Chemistry, Dr. D.Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, Maharashtra, India
| |
Collapse
|
43
|
Silva-Rudberg JA, Salardini E, O'Dell RS, Chen MK, Ra J, Georgelos JK, Morehouse MR, Melino KP, Varma P, Toyonaga T, Nabulsi NB, Huang Y, Carson RE, van Dyck CH, Mecca AP. Assessment of Gray Matter Microstructure and Synaptic Density in Alzheimer's Disease: A Multimodal Imaging Study With DTI and SV2A PET. Am J Geriatr Psychiatry 2024; 32:17-28. [PMID: 37673749 PMCID: PMC10840732 DOI: 10.1016/j.jagp.2023.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/19/2023] [Accepted: 08/05/2023] [Indexed: 09/08/2023]
Abstract
OBJECTIVE Multimodal imaging techniques have furthered our understanding of how different aspects of Alzheimer's disease (AD) pathology relate to one another. Diffusion tensor imaging (DTI) measures such as mean diffusivity (MD) may be a surrogate measure of the changes in gray matter structure associated with AD. Positron emission tomography (PET) imaging of synaptic vesicle glycoprotein 2A (SV2A) has been used to quantify synaptic loss, which is the major pathological correlate of cognitive impairment in AD. In this study, we investigated the relationship between gray matter microstructure and synaptic density. METHODS DTI was used to measure MD and [11C]UCB-J PET to measure synaptic density in 33 amyloid-positive participants with AD and 17 amyloid-negative cognitively normal (CN) participants aged 50-83. Univariate regression analyses were used to assess the association between synaptic density and MD in both the AD and CN groups. RESULTS Hippocampal MD was inversely associated with hippocampal synaptic density in participants with AD (r = -0.55, p <0.001, df = 31) but not CN (r = 0.13, p = 0.62, df = 15). Exploratory analyses across other regions known to be affected in AD suggested widespread inverse associations between synaptic density and MD in the AD group. CONCLUSION In the setting of AD, an increase in gray matter MD is inversely associated with synaptic density. These co-occurring changes may suggest a link between synaptic loss and gray matter microstructural changes in AD. Imaging studies of gray matter microstructure and synaptic density may allow important insights into AD-related neuropathology.
Collapse
Affiliation(s)
- Jason A Silva-Rudberg
- Alzheimer's Disease Research Unit (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT; Department of Psychiatry (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT.
| | - Elaheh Salardini
- Alzheimer's Disease Research Unit (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT; Department of Psychiatry (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT
| | - Ryan S O'Dell
- Alzheimer's Disease Research Unit (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT; Department of Psychiatry (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT
| | - Ming-Kai Chen
- Department of Radiology and Biomedical Imaging (M-KC, PV, TT, NBN, YH, REC), Yale University School of Medicine, New Haven, CT
| | - Jocelyn Ra
- Alzheimer's Disease Research Unit (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT; Department of Psychiatry (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT
| | - Jamie K Georgelos
- Alzheimer's Disease Research Unit (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT; Department of Psychiatry (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT
| | - Mackenzie R Morehouse
- Alzheimer's Disease Research Unit (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT; Department of Psychiatry (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT
| | - Kaitlyn P Melino
- Alzheimer's Disease Research Unit (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT; Department of Psychiatry (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT
| | - Pradeep Varma
- Department of Radiology and Biomedical Imaging (M-KC, PV, TT, NBN, YH, REC), Yale University School of Medicine, New Haven, CT
| | - Takuya Toyonaga
- Department of Radiology and Biomedical Imaging (M-KC, PV, TT, NBN, YH, REC), Yale University School of Medicine, New Haven, CT
| | - Nabeel B Nabulsi
- Department of Radiology and Biomedical Imaging (M-KC, PV, TT, NBN, YH, REC), Yale University School of Medicine, New Haven, CT
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging (M-KC, PV, TT, NBN, YH, REC), Yale University School of Medicine, New Haven, CT
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging (M-KC, PV, TT, NBN, YH, REC), Yale University School of Medicine, New Haven, CT
| | - Christopher H van Dyck
- Alzheimer's Disease Research Unit (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT; Department of Psychiatry (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT; Department of Neuroscience (CHvD), Yale University School of Medicine, New Haven, CT; Department of Neurology (CHvD), Yale University School of Medicine, New Haven, CT
| | - Adam P Mecca
- Alzheimer's Disease Research Unit (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT; Department of Psychiatry (JAS-R, ES, RSO, JR, JKG, MRM, KPM, CHvD, APM), Yale University School of Medicine, New Haven, CT.
| |
Collapse
|
44
|
Byeon G, Byun MS, Yi D, Ahn H, Jung G, Lee YS, Kim YK, Kang KM, Sohn CH, Lee DY. Moderation of Amyloid-β Deposition on the Effect of Cholinesterase Inhibitors on Cognition in Mild Cognitive Impairment. J Alzheimers Dis 2024; 101:91-97. [PMID: 39121119 PMCID: PMC11380220 DOI: 10.3233/jad-240380] [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: 08/11/2024]
Abstract
Background Clinical trial findings on cholinesterase inhibitors (ChEIs) for mild cognitive impairment (MCI) are inconclusive, offering limited support for their MCI treatment. Given that nearly half of amnestic MCI cases lack cerebral amyloid-β (Aβ) deposition, a hallmark of Alzheimer's disease; this Aβ heterogeneity may explain inconsistent results. Objective This study aimed to assess whether Aβ deposition moderates ChEI effects on amnestic MCI cognition. Methods We examined 118 individuals with amnestic MCI (ages 55-90) in a longitudinal cohort study. Baseline and 2-year follow-up assessments included clinical evaluations, neuropsychological testing, and multimodal neuroimaging. Generalized linear models were primarily analyzed to test amyloid positivity's moderation of ChEI effects on cognitive change over 2 years. Cognitive outcomes included Mini-Mental Status Examination score, the total score of the Consortium to Establish a Registry for Alzheimer's Disease neuropsychological battery, and Clinical Dementia Rating-sum of boxes. Results The analysis found no significant ChEI use x amyloid positivity interaction for all cognitive outcomes. ChEI use, irrespective of Aβ status, was associated with more cognitive decline over the 2-year period. Conclusions Aβ pathology does not appear to moderate ChEI effects on cognitive decline in MCI.
Collapse
Affiliation(s)
- Gihwan Byeon
- Department of Neuropsychiatry, Kangwon National University Hospital, Chuncheon, Korea
| | - Min Soo Byun
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Korea
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, Korea
| | - Hyejin Ahn
- Interdisciplinary Program of Cognitive Science, Seoul National University College of Humanities, Seoul, Korea
| | - Gijung Jung
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, Korea
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, SMG- SNU Boramae Medical Center, Seoul, Korea
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, Seoul, Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, Seoul, Korea
| | - Dong Young Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Korea
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Korea
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, Korea
- Interdisciplinary Program of Cognitive Science, Seoul National University College of Humanities, Seoul, Korea
| |
Collapse
|
45
|
Kordi MR, Khademi N, Zobeydi AM, Torabi S, Mahmoodifar E, Gaeini AA, Choobineh S, Pournemati P. High-intensity interval training combined with cannabidiol supplementation improves cognitive impairment by regulating the expression of apolipoprotein E, presenilin-1, and glutamate proteins in a rat model of amyloid β-induced Alzheimer's disease. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:1583-1591. [PMID: 39539449 PMCID: PMC11556759 DOI: 10.22038/ijbms.2024.79464.17210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 06/16/2024] [Indexed: 11/16/2024]
Abstract
Objectives Alzheimer's disease (AD) is a major public concern and one of the primary types of dementia characterized by memory impairment and cognitive decline. Although the properties of exercise training and cannabidiol (CBD) treatments for improving AD have recently been revealed, the exact mechanisms remain unknown. Therefore, this study highlights the interactive impact of high-intensity interval training (HIIT) and CBD administration on improving cognitive impairment in a rat model of amyloid beta (Aꞵ)-induced AD through modulating the expression of apolipoprotein E (APOE), presenilin-1, and glutamate proteins. Materials and Methods After acclimatization, the animals were randomly assigned into seven subgroups: control (CNT), Sham, Alzheimer (AL), Alzheimer + HIIT (AL + HIIT), Alzheimer + cannabidiol (AL + CBD), Alzheimer + CBD + HIIT (AL + CBD + HIIT), and model (sacrificed ten days after surgery to confirm the induction of AD) groups. To induce AD, rats received an intrahippocampal injection of Aꞵ. The animals in exercise groups performed the HIIT protocol, and the rats in CBD groups were administered 20 mg/kg CBD suspended in sesame oil for six weeks. Following the experimental protocol, serum and hippocampus tissue were collected for histopathological and western blot analysis. Results Our findings indicated that both HIIT and CBD treatments were efficacious in ameliorating Aꞵ deposition and modulating biomarkers of AD, including APOE, presenilin-1, and glutamate. However, the interactive effect of HIIT and CBD supplementation was more effective. Conclusion Our findings demonstrated the positive therapeutic effect of HIIT and CBD interventions, particularly HIIT combined with CBD, on alleviating AD.
Collapse
Affiliation(s)
- Mohammad Reza Kordi
- Department of Exercise Physiology, Faculty of Sports Science and Health, University of Tehran, Tehran, Iran
| | - Nooshin Khademi
- Department of Exercise Physiology, Faculty of Sports Science and Health, University of Tehran, Tehran, Iran
| | - Amir Mohammad Zobeydi
- Department of Exercise Physiology, Faculty of Sports Science and Health, University of Tehran, Tehran, Iran
| | - Samane Torabi
- Department of Exercise Physiology, Faculty of Sports Science and Health, University of Tehran, Tehran, Iran
| | - Esmaeil Mahmoodifar
- Department of Exercise Physiology, Faculty of Sports Science and Health, University of Tehran, Tehran, Iran
| | - Abbas Ali Gaeini
- Department of Exercise Physiology, Faculty of Sports Science and Health, University of Tehran, Tehran, Iran
| | - Siroos Choobineh
- Department of Exercise Physiology, Faculty of Sports Science and Health, University of Tehran, Tehran, Iran
| | - Parisa Pournemati
- Department of Exercise Physiology, Faculty of Sports Science and Health, University of Tehran, Tehran, Iran
| |
Collapse
|
46
|
Han SM, Byun MS, Yi D, Jung JH, Kong N, Chang YY, Keum M, Jung GJ, Lee JY, Lee YS, Kim YK, Kang KM, Sohn CH, Lee DY. Modulatory Effect of Blood LDL Cholesterol on the Association between Cerebral Aβ and Tau Deposition in Older Adults. J Prev Alzheimers Dis 2024; 11:1767-1774. [PMID: 39559888 PMCID: PMC11573824 DOI: 10.14283/jpad.2024.131] [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: 11/20/2024]
Abstract
BACKGROUND This study investigates the synergistic relationship between blood low-density lipoprotein cholesterol (LDL-C) and cerebral beta-amyloid (Aβ) in relation to tau deposition, a key factor in the pathology of Alzheimer's disease (AD), in older adults across a diverse cognitive spectrum. OBJECTIVES To examine whether higher levels of LDL-C in the blood moderate the association of cerebral Aβ with tau deposition in older adults, including those with normal cognition, mild cognitive impairment, and Alzheimer's disease dementia. DESIGN Cross-sectional design. SETTING The study was conducted as a part of a prospective cohort study. All assessments were done at the Seoul National University Hospital, Seoul, South Korea. PARTICIPANTS A total of 136 older adults (aged 60-85 years) with normal cognition, mild cognitive impairment or Alzheimer's disease (AD) dementia were included. MEASUREMENTS Serum lipid measurements, [11C] Pittsburgh Compound B-positron emission tomography (PET), [18F] AV-1451 PET, and magnetic resonance imaging were performed on all participants. RESULTS There was a significant Aβ x LDL-C interaction effect on tau deposition indicating a synergistic moderation effect of LDL-C on the relationship between Aβ and tau deposition. Subsequent subgroup analysis showed that the positive association between Aβ and tau deposition was stronger in higher LDL-C group than in lower LDL-C group. In contrast, other lipids, such as total cholesterol, high-density lipoprotein cholesterol, and triglycerides, did not show a similar moderation effect on the relationship between Aβ deposition and tau deposition. CONCLUSION Our findings suggest that blood LDL-C synergistically enhances the influence of Aβ deposition on tau pathology, emphasizing the need for greater attention to the role of LDL-C in AD progression.
Collapse
Affiliation(s)
- S M Han
- Dong Young Lee, Department of Neuropsychiatry, Seoul National University Hospital, 101, Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea; Telephone: +82-2-2072-2205, Fax: +82-2-744-2471,
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
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
|
48
|
Bai H, Naj AC, Benchek P, Dumitrescu L, Hohman T, Hamilton-Nelson K, Kallianpur AR, Griswold AJ, Vardarajan B, Martin ER, Beecham GW, Below JE, Schellenberg G, Mayeux R, Farrer L, Pericak-Vance MA, Haines JL, Bush WS. A haptoglobin (HP) structural variant alters the effect of APOE alleles on Alzheimer's disease. Alzheimers Dement 2023; 19:4886-4895. [PMID: 37051669 DOI: 10.1002/alz.13050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 04/14/2023]
Abstract
BACKGROUND Haptoglobin (HP) is an antioxidant of apolipoprotein E (APOE), and previous reports have shown HP binds with APOE and amyloid beta (Aβ) to aid its clearance. A common structural variant of the HP gene distinguishes it into two alleles: HP1 and HP2. METHODS HP genotypes were imputed in 29 cohorts from the Alzheimer's Disease Genetics Consortium (N = 20,512). Associations between the HP polymorphism and Alzheimer's disease (AD) risk and age of onset through APOE interactions were investigated using regression models. RESULTS The HP polymorphism significantly impacts AD risk in European-descent individuals (and in meta-analysis with African-descent individuals) by modifying both the protective effect of APOE ε2 and the detrimental effect of APOE ε4. The effect is particularly significant among APOE ε4 carriers. DISCUSSION The effect modification of APOE by HP suggests adjustment and/or stratification by HP genotype is warranted when APOE risk is considered. Our findings also provided directions for further investigations on potential mechanisms behind this association.
Collapse
Affiliation(s)
- Haimeng Bai
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA
- Systems Biology and Bioinformatics, Department of Nutrition, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Adam C Naj
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Penelope Benchek
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Population & Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Logan Dumitrescu
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Timothy Hohman
- Vanderbilt Memory & Alzheimer's Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kara Hamilton-Nelson
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, USA
| | - Asha R Kallianpur
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
- Department of Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Anthony J Griswold
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, USA
| | - Badri Vardarajan
- Department of Neurology, The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, USA
| | - Eden R Martin
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, USA
| | - Gary W Beecham
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, USA
| | - Jennifer E Below
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Gerard Schellenberg
- Penn Neurodegeneration Genomics Center, Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Richard Mayeux
- Department of Neurology, The Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, USA
| | - Lindsay Farrer
- Departments of Medicine (Biomedical Genetics), Neurology, Ophthalmology, Biostatistics, and Epidemiology, Boston University Schools of Medicine and Public Health, Boston, Massachusetts, USA
| | | | - Jonathan L Haines
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Population & Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - William S Bush
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Population & Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
49
|
Rasmussen KL, Luo J, Nordestgaard BG, Tybjærg-Hansen A, Frikke-Schmidt R. APOE and vascular disease: Sequencing and genotyping in general population cohorts. Atherosclerosis 2023; 385:117218. [PMID: 37586954 DOI: 10.1016/j.atherosclerosis.2023.117218] [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: 04/14/2023] [Revised: 07/14/2023] [Accepted: 08/08/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND AND AIMS The apolipoprotein E(APOE) ϵ2/ϵ3/ϵ4 polymorphism plays a central role in lipid metabolism, vascular disease and dementia. The impact of the full range of structural genetic variation in APOE for lipids, lipoproteins and apolipoproteins and for vascular disease in the general population is not known. METHODS We systematically sequenced APOE in 10,296 individuals from the Copenhagen City Heart Study and genotyped nine rare variants (frequency≥2/10,296) in 95,227 individuals from the Copenhagen General Population Study. The UK Biobank was used for validation of common APOE variants. RESULTS Rare mutations in APOE, predicted to be deleterious, are present in 1 in 257 individuals in the general population. In the meta-analysis, multifactorially adjusted hazard ratios (95% confidence intervals) for ϵ44 and ϵ22 versus ϵ33 were 1.15 (1.04-1.26) and 1.02 (0.83-1.24) for ischemic cerebrovascular disease (ICVD), 1.11 (1.04-1.19) and 0.94 (0.83-1.08) for ischemic heart disease (IHD) and 1.03 (0.89-1.17) and 1.49 (1.20-1.87) for peripheral arterial disease (PAD). A multifactorially and ϵ2/ϵ3/ϵ4 adjusted weighted allele score on the continuous scale including all common and rare structural variants showed that for individuals with genetically predicted high plasma apoE and remnant cholesterol the risk for PAD was increased. CONCLUSIONS APOE variants with high apoE, triglycerides, and remnant cholesterol are associated with PAD, whereas common APOE variants with high LDL cholesterol, triglycerides and remnant cholesterol are associated with IHD. APOE variants with low apoE are associated with increased risk of ICVD. These findings highlight that both rare and common structural variations in APOE play a role in vascular disease.
Collapse
Affiliation(s)
- Katrine L Rasmussen
- Department of Clinical Biochemistry, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark; The Copenhagen General Population Study, Herlev and Gentofte Hospital, Herlev Ringvej 75, DK-2730 Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen, Denmark; Department of Clinical Biochemistry, Nordsjællands Hospital, Dyrehavevej 29, DK-3400 Hillerød, Denmark.
| | - Jiao Luo
- Department of Clinical Biochemistry, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark; The Copenhagen General Population Study, Herlev and Gentofte Hospital, Herlev Ringvej 75, DK-2730 Herlev, Denmark
| | - Børge G Nordestgaard
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Herlev Ringvej 75, DK-2730 Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen, Denmark; The Copenhagen City Heart Study, Frederiksberg Hospital, Nordre Fasanvej 57, DK-2000 Frederiksberg, Denmark; Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Herlev Ringvej 75, DK-2730 Herlev, Denmark
| | - Anne Tybjærg-Hansen
- Department of Clinical Biochemistry, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark; The Copenhagen General Population Study, Herlev and Gentofte Hospital, Herlev Ringvej 75, DK-2730 Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen, Denmark; The Copenhagen City Heart Study, Frederiksberg Hospital, Nordre Fasanvej 57, DK-2000 Frederiksberg, Denmark
| | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark; The Copenhagen General Population Study, Herlev and Gentofte Hospital, Herlev Ringvej 75, DK-2730 Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen, Denmark
| |
Collapse
|
50
|
Palmer JM, Huentelman M, Ryan L. More than just risk for Alzheimer's disease: APOE ε4's impact on the aging brain. Trends Neurosci 2023; 46:750-763. [PMID: 37460334 DOI: 10.1016/j.tins.2023.06.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/16/2023] [Accepted: 06/12/2023] [Indexed: 08/18/2023]
Abstract
The apolipoprotein ε4 (APOE ε4) allele is most commonly associated with increased risk for late-onset Alzheimer's disease (AD). However, recent longitudinal studies suggest that these risks are overestimated; most ε4 carriers will not develop dementia in their lifetime. In this article, we review new evidence regarding the impact of APOE ε4 on cognition among healthy older adults. We discuss emerging work from animal models suggesting that ε4 impacts brain structure and function in multiple ways that may lead to age-related cognitive impairment, independent from AD pathology. We discuss the importance of taking an individualized approach in future studies by incorporating biomarkers and neuroimaging methods that may better disentangle the phenotypic influences of APOE ε4 on the aging brain from prodromal AD pathology.
Collapse
Affiliation(s)
- Justin M Palmer
- The University of Arizona, Tucson, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA.
| | - Matthew Huentelman
- Translational Genomics Research Institute, Phoenix, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Lee Ryan
- The University of Arizona, Tucson, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA.
| |
Collapse
|