1
|
Shvetcov A, Thomson S, Cho A, Wilkins HM, Reed JH, Swerdlow RH, Brown DA, Finney CA. Proteome profiling of cerebrospinal fluid using machine learning shows a unique protein signature associated with APOE4 genotype. Aging Cell 2025; 24:e14439. [PMID: 39722190 PMCID: PMC11984689 DOI: 10.1111/acel.14439] [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: 06/16/2024] [Revised: 11/07/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
Proteome changes associated with APOE4 variant carriage that are independent of Alzheimer's disease (AD) pathology and diagnosis are unknown. This study investigated APOE4 proteome changes in people with AD, mild cognitive impairment, and no impairment. Clinical, APOE genotype, and cerebrospinal fluid (CSF) proteome and AD biomarker data was sourced from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database. Proteome profiling was done using supervised machine learning. We found an APOE4-specific proteome signature that was independent of cognitive diagnosis and AD pathological biomarkers, and increased the risk of progression to cognitive impairment. Proteins were enriched in brain regions including the caudate and cortex and cells including endothelial cells, oligodendrocytes, and astrocytes. Enriched peripheral immune cells included T cells, macrophages, and B cells. APOE4 carriers have a unique CSF proteome signature associated with a strong brain and peripheral immune and inflammatory phenotype that likely underlies APOE4 carriers' vulnerability to cognitive decline and AD as they age.
Collapse
Affiliation(s)
- Artur Shvetcov
- Translational Dementia Research Group, Centre for Immunology and Allergy ResearchWestmead Institute for Medical ResearchSydneyNSWAustralia
- Department of Psychological MedicineSydney Children's Hospital NetworkSydneyNSWAustralia
- Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and HealthUniversity of New South WalesSydneyNSWAustralia
| | - Shannon Thomson
- Translational Dementia Research Group, Centre for Immunology and Allergy ResearchWestmead Institute for Medical ResearchSydneyNSWAustralia
- School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneySydneyNSWAustralia
| | - Ann‐Na Cho
- Human Brain Microphysiology Systems Group, School of Biomedical Engineering, Faculty of EngineeringThe University of SydneySydneyNSWAustralia
| | - Heather M. Wilkins
- University of Kansas Alzheimer's Disease Research CentreKansas CityKSUSA
- Department of Biochemistry and Molecular BiologyUniversity of Kansas Medical CentreKansas CityKSUSA
- Department of NeurologyUniversity of Kansas Medical CentreKansas CityKSUSA
| | - Joanne H. Reed
- School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneySydneyNSWAustralia
- Autoimmunity and Amyloidosis Research Group, Centre for Immunology and Allergy ResearchWestmead Institute for Medical ResearchSydneyNSWAustralia
| | - Russell H. Swerdlow
- University of Kansas Alzheimer's Disease Research CentreKansas CityKSUSA
- Department of Biochemistry and Molecular BiologyUniversity of Kansas Medical CentreKansas CityKSUSA
- Department of NeurologyUniversity of Kansas Medical CentreKansas CityKSUSA
- Department of Molecular and Integrative PhysiologyUniversity of Kansas Medical CentreKansas CityKSUSA
| | - David A. Brown
- Neuroinflammation Research Group, Centre for Immunology and Allergy ResearchWestmead Institute for Medical ResearchSydneyNSWAustralia
- Department of ImmunopathologyInstitute for Clinical Pathology and Medical Research‐New South Wales Health PathologySydneyNSWAustralia
- Westmead Clinical School, Faculty of Medicine and HealthThe University of SydneySydneyNSWAustralia
| | | | - Caitlin A. Finney
- Translational Dementia Research Group, Centre for Immunology and Allergy ResearchWestmead Institute for Medical ResearchSydneyNSWAustralia
- School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneySydneyNSWAustralia
| |
Collapse
|
2
|
Feng Y, Laraib A, Lin X, Li Q, Zhan J, Li X. Associations of tau, Aβ, and brain volume of the Papez circuit with cognition in Alzheimer's disease. Eur Arch Psychiatry Clin Neurosci 2025; 275:241-256. [PMID: 38824476 DOI: 10.1007/s00406-024-01827-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 05/10/2024] [Indexed: 06/03/2024]
Abstract
This study aimed to investigate the cross-sectional associations between regional Alzheimer's disease (AD) biomarkers, including tau, β-amyloid (Aβ), and brain volume, within the Papez circuit, and neuropsychological functioning across the preclinical and clinical spectrum of AD. We utilized data from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database, including 251 Aβ-positive participants. Participants were categorized into three groups based on the Clinical Dementia Rating (CDR): 73 individuals with preclinical AD (CDR = 0), 114 with prodromal AD (CDR = 0.5), and 64 with clinical AD dementia (CDR ≥ 1). Linear regression analyses, adjusted for age, gender, and education years, were employed to evaluate the associations between five regions of interest (the hippocampus, para-hippocampus, entorhinal cortex, posterior cingulate cortex, and thalamus) and five neuropsychological tests across the three imaging modalities. In the preclinical stage of AD, flortaucipir PET was associated with impaired global cognition and episodic memory (range standardized β = 0.255-0.498, p < 0.05 corrected for multiple comparisons), while florbetapir PET and brain volume were marginally related to global cognition (range standardized β = 0.221-0.231, p < 0.05). In the clinical stages of AD (prodromal and dementia), both increased flortaucipir uptake and decreased brain volume were significantly associated with poorer global neuropsychological and episodic memory performance (range standardized β = 0.222-0.621, p < 0.05, most regions of interest survived correction for multiple comparisions). However, a slight relationship was observed between florbetapir uptake and poorer global cognitive function. The regions most affected by flortaucipir PET were the hippocampus, para-hippocampus, and posterior cingulate cortex. During the clinical stages, the hippocampus and entorhinal cortex exhibited the most significant volumetric changes. Tau PET and brain volume measurements within the Papez circuit are more sensitive indicators of early cognitive deficits in AD than Aβ PET. Furthermore, during the clinical stages of AD, both flortaucipir PET and brain volume of the Papez circuit are closely correlated with cognitive decline. These findings underscore the importance of integrating multiple biomarkers for the comprehensive evaluation of AD pathology and its impact on cognition.
Collapse
Affiliation(s)
- Yuxue Feng
- Department of Neurology, The Fifth People's Hospital of Chongqing, Chongqing, China
| | - Azka Laraib
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74-76 Linjiang Road, Yuzhong District, Chongqing, 400000, China
| | - Xiuqi Lin
- Chongqing Medical University, Chongqing, China
| | - Qin Li
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74-76 Linjiang Road, Yuzhong District, Chongqing, 400000, China
| | - Jiehong Zhan
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74-76 Linjiang Road, Yuzhong District, Chongqing, 400000, China
| | - Xiaofeng Li
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74-76 Linjiang Road, Yuzhong District, Chongqing, 400000, China.
- Department of Neurology, People's Hospital of Linshui County, Guangan, China.
| |
Collapse
|
3
|
Chaloemtoem A, Thornton V, Chang Y, Anokhin AP, Belloy ME, Bijsterbosch J, Gordon BA, Hartz SM, Bierut LJ. Hippocampal volumes in UK Biobank are associated with APOE only in older adults. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e70024. [PMID: 39553251 PMCID: PMC11567847 DOI: 10.1002/dad2.70024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/04/2024] [Accepted: 09/17/2024] [Indexed: 11/19/2024]
Abstract
INTRODUCTION The hippocampus atrophies with age and is implicated in neurodegenerative disorders including Alzheimer's disease (AD). We examined the interplay between age and apolipoprotein E (APOE) genotype on total hippocampal volume. METHODS Using neuroimaging data from 37,463 UK Biobank participants, we applied linear regression to quantify the association of age and APOE with hippocampal volume and identified the age when volumes of ε2/ε3, ε3/ε4, and ε4/ε4 carriers significantly deviated from ε3/ε3 using generalized additive modeling. RESULTS Total hippocampal volume declined with age, with significant differences by APOE genotype emerging after age 60. ε3/ε4 and ε4/ε4 carriers displayed reduced volumes from ages 69 and 61, respectively, while ε2/ε3 showed delayed decline starting at the age of 76. DISCUSSION The association of APOE and hippocampal volume is age-dependent, with differences in volumes of ε4/ε4 carriers detected as early as age 61. This work underscores the importance of APOE genotype in determining when to begin screening for AD. Highlights Apolipoprotein E (APOE) genotype shows an age-dependent association with total hippocampal volume.No association between APOE and total hippocampal volume was detected before age 60.Accelerated decline was observed in ε4/ε4 carriers at age 61 and ε3/ε4 at age 69.Delayed decline was evident in ε2/ε3 carriers starting at age 76.
Collapse
Affiliation(s)
- Ariya Chaloemtoem
- Department of PsychiatryWashington University School of MedicineSt. LouisMissouriUSA
| | - Vera Thornton
- Department of PsychiatryWashington University School of MedicineSt. LouisMissouriUSA
| | - Yoonhoo Chang
- Department of PsychiatryWashington University School of MedicineSt. LouisMissouriUSA
| | - Andrey P. Anokhin
- Department of PsychiatryWashington University School of MedicineSt. LouisMissouriUSA
| | - Michaël E. Belloy
- NeuroGenomics and Informatics CenterWashington University School of MedicineSt. LouisMissouriUSA
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| | - Janine Bijsterbosch
- Department of RadiologyWashington University School of MedicineSt. LouisMissouriUSA
| | - Brian A. Gordon
- NeuroGenomics and Informatics CenterWashington University School of MedicineSt. LouisMissouriUSA
- Department of RadiologyWashington University School of MedicineSt. LouisMissouriUSA
| | - Sarah M. Hartz
- Department of PsychiatryWashington University School of MedicineSt. LouisMissouriUSA
| | - Laura J. Bierut
- Department of PsychiatryWashington University School of MedicineSt. LouisMissouriUSA
| |
Collapse
|
4
|
Muñoz-Neira C, Zeng J, Kucikova L, Huang W, Xiong X, Muniz-Terrera G, Ritchie C, O'Brien JT, Su L. Differences in Grey Matter Concentrations and Functional Connectivity between Young Carriers and Non-Carriers of the APOE ε4 Genotype. J Clin Med 2024; 13:5228. [PMID: 39274441 PMCID: PMC11396314 DOI: 10.3390/jcm13175228] [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: 07/18/2024] [Revised: 08/24/2024] [Accepted: 08/30/2024] [Indexed: 09/16/2024] Open
Abstract
Background: The pathophysiology of Alzheimer's disease (AD) may begin developing years or even decades prior to the manifestation of its first symptoms. The APOE ε4 genotype is a prominent genetic risk for AD that has been found to be associated with brain changes across the lifespan since early adulthood. Thus, studying brain changes that may occur in young adults with an APOE ε4 status is highly relevant. Objective: Examine potential differences in grey matter (GM) and functional connectivity (FC) in brains of cognitively healthy young APOE ε4 carriers and non-carriers, denoted here as ε4(+) and ε4(-), respectively. Methods: Three Tesla magnetic resonance imaging (MRI) brain scans were acquired from cognitively healthy young participants aged approximately 20 years (n = 151). Voxel-based morphometry (VBM) analysis was employed to identify potential structural differences in GM between ε4(+) and ε4(-). In a subsequent seed-based connectivity (SBC) analysis, brain regions that structurally differed in the VBM analysis were considered as seeds and correlated with all the remaining voxels across the brains to then measure the differences in FC between groups. Results: The VBM analysis suggested that ε4(+) (n = 28) had greater GM densities relative to ε4(-) (n = 123) in the left hippocampus and the left posterior insula (puncorr < 0.001). However, the effect did not survive the correction for multiple comparisons, suggesting minimal structural differences in this age range. In contrast, the SBC analysis indicated that ε4(+) exhibited significantly decreased FC between the left hippocampus and areas of the left middle temporal gyrus (n = 27) compared to ε4(-) (n = 102). These results remained significant after multiple comparisons (pFDR < 0.05). Lastly, no statistically significant differences in FC between groups were observed for the left insular seed (pFDR > 0.05). Discussion: These results suggest early structural and functional brain changes associated with the APOE ε4 genotype on young adults. Yet, they must be cautiously interpreted and contrasted with both older adults with genetic risk for AD and patients diagnosed with AD.
Collapse
Affiliation(s)
- Carlos Muñoz-Neira
- Artificial Intelligence & Computational Neuroscience Group (AICN Group), Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield S10 2HQ, UK
- Old Age Psychiatry Research Group (OAP Group), Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Jianmin Zeng
- Sino-Britain Centre for Cognition and Ageing Research, Faculty of Psychology, Southwest University, Chongqing 400715, China
| | - Ludmila Kucikova
- Artificial Intelligence & Computational Neuroscience Group (AICN Group), Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield S10 2HQ, UK
- Insigneo Institute for In Silico Medicine, University of Sheffield, Sheffield S1 3JD, UK
| | - Weijie Huang
- Artificial Intelligence & Computational Neuroscience Group (AICN Group), Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield S10 2HQ, UK
- Old Age Psychiatry Research Group (OAP Group), Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SZ, UK
- School of Systems Science, Beijing Normal University, Beijing 100875, China
| | - Xiong Xiong
- Artificial Intelligence & Computational Neuroscience Group (AICN Group), Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield S10 2HQ, UK
- School of Information and Communication Engineering, Beijing University of Posts and Telecommunications, Beijing 100876, China
| | - Graciela Muniz-Terrera
- Edinburgh Dementia Prevention, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH4 2XU, UK
- Ohio University Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Craig Ritchie
- Edinburgh Dementia Prevention, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH4 2XU, UK
- Scottish Brain Sciences, Edinburgh EH12 9DQ, UK
| | - John T O'Brien
- Old Age Psychiatry Research Group (OAP Group), Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Li Su
- Artificial Intelligence & Computational Neuroscience Group (AICN Group), Sheffield Institute for Translational Neuroscience (SITraN), Division of Neuroscience, School of Medicine and Population Health, Faculty of Health, University of Sheffield, Sheffield S10 2HQ, UK
- Old Age Psychiatry Research Group (OAP Group), Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SZ, UK
- Insigneo Institute for In Silico Medicine, University of Sheffield, Sheffield S1 3JD, UK
| |
Collapse
|
5
|
Pehlivanoglu D, Shoenfelt A, Hakim Z, Heemskerk A, Zhen J, Mosqueda M, Wilson RC, Huentelman M, Grilli MD, Turner G, Spreng RN, Ebner NC. Phishing vulnerability compounded by older age, apolipoprotein E e4 genotype, and lower cognition. PNAS NEXUS 2024; 3:pgae296. [PMID: 39118834 PMCID: PMC11309394 DOI: 10.1093/pnasnexus/pgae296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024]
Abstract
With technological advancements, financial exploitation tactics have expanded into the online realm. Older adults may be particularly susceptible to online scams due to age- and Alzheimer's disease-related changes in cognition. In this study, 182 adults ranging from 18 to 90 years underwent cognitive assessment, genotyping for apolipoprotein E e4 (APOE4), and completed the lab-based Short Phishing Email Suspicion Test (S-PEST) as well as the real-life PHishing Internet Task (PHIT). Across both paradigms, older age predicted heightened susceptibility to phishing, with this enhanced susceptibility pronounced among older APOE4 allele carriers with lower working memory. Additionally, performance in both phishing tasks was correlated in that reduced ability to discriminate between phishing and safe emails in S-PEST predicted greater phishing susceptibility in PHIT. The current study identifies older age, APOE4, and lower cognition as risk factors for phishing vulnerability and introduces S-PEST as an easy-to-administer, ecologically valid tool for assessing phishing susceptibility.
Collapse
Affiliation(s)
- Didem Pehlivanoglu
- Department of Psychology, University of Florida, 945 Center Dr, Gainesville, FL 32603, USA
- Florida Institute for National Security, University of Florida, 601 Gale Lemerand Dr, Gainesville, FL 32611, USA
| | - Alayna Shoenfelt
- Department of Psychology, University of Florida, 945 Center Dr, Gainesville, FL 32603, USA
| | - Ziad Hakim
- Department of Psychology, University of Florida, 945 Center Dr, Gainesville, FL 32603, USA
| | - Amber Heemskerk
- Department of Psychology, University of Florida, 945 Center Dr, Gainesville, FL 32603, USA
| | - Jialong Zhen
- Department of Psychology, University of Florida, 945 Center Dr, Gainesville, FL 32603, USA
| | - Mario Mosqueda
- Translational Genomics Research Institute, 445 N 5th St 4th Floor, Phoenix, AZ 85004, USA
| | - Robert C Wilson
- Department of Psychology, University of Arizona, 1503 E. University Blvd., Tucson, AZ 85721, USA
| | - Matthew Huentelman
- Translational Genomics Research Institute, 445 N 5th St 4th Floor, Phoenix, AZ 85004, USA
| | - Matthew D Grilli
- Department of Psychology, University of Arizona, 1503 E. University Blvd., Tucson, AZ 85721, USA
| | - Gary Turner
- Department of Psychology, York University, 4700 Keele St, North York, ON M3J 1P3, Canada
| | - R Nathan Spreng
- Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue West, Montreal, QC H3A 1A1, Canada
| | - Natalie C Ebner
- Department of Psychology, University of Florida, 945 Center Dr, Gainesville, FL 32603, USA
- Florida Institute for National Security, University of Florida, 601 Gale Lemerand Dr, Gainesville, FL 32611, USA
- Florida Institute for Cybersecurity Research, University of Florida, Malachowsky Hall, 1889 Museum Rd, Gainesville, FL 32603, USA
- McKnight Brain Institute, University of Florida, 1149 Newell Dr, Gainesville, FL 32610, USA
| |
Collapse
|
6
|
Popov VA, Ukraintseva SV, Duan H, Yashin AI, Arbeev KG. Traffic-related air pollution and APOE4 can synergistically affect hippocampal volume in older women: new findings from UK Biobank. FRONTIERS IN DEMENTIA 2024; 3:1402091. [PMID: 39135618 PMCID: PMC11317402 DOI: 10.3389/frdem.2024.1402091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 07/10/2024] [Indexed: 08/15/2024]
Abstract
A growing research body supports the connection between neurodegenerative disorders, including Alzheimer's disease (AD), and traffic-related air pollution (TRAP). However, the underlying mechanisms are not well understood. A deeper investigation of TRAP effects on hippocampal volume (HV), a major biomarker of neurodegeneration, may help clarify these mechanisms. Here, we explored TRAP associations with the HV in older participants of the UK Biobank (UKB), taking into account the presence of APOE e4 allele (APOE4), the strongest genetic risk factor for AD. Exposure to TRAP was approximated by the distance of the participant's main residence to the nearest major road (DNMR). The left/right HV was measured by magnetic resonance imaging (MRI) in cubic millimeters (mm3). Analysis of variance (ANOVA), Welch test, and regression were used to examine statistical significance. We found significant interactions between DNMR and APOE4 that influenced HV. Specifically, DNMR <50m (equivalent of a chronically high exposure to TRAP), and carrying APOE4 were synergistically associated with a significant (P = 0.01) reduction in the right HV by about 2.5% in women aged 60-75 years (results for men didn't reach a statistical significance). Results of our study suggest that TRAP and APOE4 jointly promote neurodegeneration in women. Living farther from major roads may help reduce the risks of neurodegenerative disorders, including AD, in female APOE4 carriers.
Collapse
|
7
|
Onos KD, Lin PB, Pandey RS, Persohn SA, Burton CP, Miner EW, Eldridge K, Kanyinda JN, Foley KE, Carter GW, Howell GR, Territo PR. Assessment of neurovascular uncoupling: APOE status is a key driver of early metabolic and vascular dysfunction. Alzheimers Dement 2024; 20:4951-4969. [PMID: 38713704 PMCID: PMC11247674 DOI: 10.1002/alz.13842] [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: 12/08/2023] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 05/09/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common cause of dementia worldwide, with apolipoprotein Eε4 (APOEε4) being the strongest genetic risk factor. Current clinical diagnostic imaging focuses on amyloid and tau; however, new methods are needed for earlier detection. METHODS PET imaging was used to assess metabolism-perfusion in both sexes of aging C57BL/6J, and hAPOE mice, and were verified by transcriptomics, and immunopathology. RESULTS All hAPOE strains showed AD phenotype progression by 8 months, with females exhibiting the regional changes, which correlated with GO-term enrichments for glucose metabolism, perfusion, and immunity. Uncoupling analysis revealed APOEε4/ε4 exhibited significant Type-1 uncoupling (↓ glucose uptake, ↑ perfusion) at 8 and 12 months, while APOEε3/ε4 demonstrated Type-2 uncoupling (↑ glucose uptake, ↓ perfusion), while immunopathology confirmed cell specific contributions. DISCUSSION This work highlights APOEε4 status in AD progression manifests as neurovascular uncoupling driven by immunological activation, and may serve as an early diagnostic biomarker. HIGHLIGHTS We developed a novel analytical method to analyze PET imaging of 18F-FDG and 64Cu-PTSM data in both sexes of aging C57BL/6J, and hAPOEε3/ε3, hAPOEε4/ε4, and hAPOEε3/ε4 mice to assess metabolism-perfusion profiles termed neurovascular uncoupling. This analysis revealed APOEε4/ε4 exhibited significant Type-1 uncoupling (decreased glucose uptake, increased perfusion) at 8 and 12 months, while APOEε3/ε4 demonstrated significant Type-2 uncoupling (increased glucose uptake, decreased perfusion) by 8 months which aligns with immunopathology and transcriptomic signatures. This work highlights that there may be different mechanisms underlying age related changes in APOEε4/ε4 compared with APOEε3/ε4. We predict that these changes may be driven by immunological activation and response, and may serve as an early diagnostic biomarker.
Collapse
Affiliation(s)
| | - Peter B. Lin
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Department of NeurologyWashington University in St. LouisSt. LouisMissouriUSA
| | - Ravi S. Pandey
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
| | - Scott A. Persohn
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
| | - Charles P. Burton
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
| | - Ethan W. Miner
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
| | - Kierra Eldridge
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
| | | | - Kate E. Foley
- The Jackson LaboratoryBar HarborMaineUSA
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
| | - Gregory W. Carter
- The Jackson LaboratoryBar HarborMaineUSA
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
| | | | - Paul R. Territo
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Department of MedicineDivision of Clinical PharmacologyIndiana University School of MedicineIndianapolisIndianaUSA
| |
Collapse
|
8
|
Shvetcov A, Thomson S, Cho AN, Wilkins HM, Reed JH, Swerdlow RH, Brown DA, Finney CA. Proteome profiling of cerebrospinal fluid using machine learning shows a unique protein signature associated with APOE4 genotype. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.18.590160. [PMID: 38915547 PMCID: PMC11195053 DOI: 10.1101/2024.04.18.590160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
INTRODUCTION Proteome changes associated with APOE4 variant carriage that are independent of Alzheimer's disease (AD) pathology and diagnosis are unknown. This study investigated APOE4 proteome changes in people with AD, mild cognitive impairment, and no impairment. METHODS Clinical, APOE genotype, and cerebrospinal fluid (CSF) proteome and AD biomarker data was sourced from the Alzheimer's Disease Neuroimaging Initiative (ADNI) database. Proteome profiling was done using supervised machine learning. RESULTS We found an APOE4-specific proteome signature that was independent of cognitive diagnosis and AD pathological biomarkers, and increased risk of progression to cognitive impairment. Proteins were enriched in brain regions including the caudate and cortex and cells including endothelial cells, oligodendrocytes, and astrocytes. Enriched peripheral immune cells included T cells, macrophages, and B cells. DISCUSSION APOE4 carriers have a unique CSF proteome signature associated with a strong brain and peripheral immune and inflammatory phenotype that likely underlies APOE4 carriers' vulnerability to cognitive decline and AD.
Collapse
Affiliation(s)
- Artur Shvetcov
- Department of Psychological Medicine, Sydney Children’s Hospital Network, Sydney, NSW, Australia
| | - Shannon Thomson
- Neuroinflammation Research Group, Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Ann-Na Cho
- Human Brain Microphysiology Systems Group, School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, NSW, Australia
| | - Heather M. Wilkins
- University of Kansas Alzheimer’s Disease Centre, Kansas City, KS, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Centre, Kansas City, KS, USA
- Department of Neurology, University of Kansas Medical Centre, Kansas City, KS, USA
| | - Joanne H. Reed
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Autoimmunity and Amyloidosis Research Group, Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Russell H. Swerdlow
- University of Kansas Alzheimer’s Disease Centre, Kansas City, KS, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Centre, Kansas City, KS, USA
- Department of Neurology, University of Kansas Medical Centre, Kansas City, KS, USA
- Department of Molecular and Integrative Physiology, University of Kansas Medical Centre, Kansas City, KS, USA
| | - David A. Brown
- Neuroinflammation Research Group, Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, Sydney, NSW, Australia
- Department of Immunopathology, Institute for Clinical Pathology and Medical Research-New South Wales Health Pathology, Sydney, NSW, Australia
- Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Alzheimer’s Disease Neuroimaging Initiative
- Data used in preparation of this article were obtained from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) database (adni.loni.usc.edu). As such, the investigators within the ADNI contributed to the design and implementation of ADNI and/or provided data but did not participate in analysis or writing of this report. A complete listing of ADNI investigators can be found at: http://adni.loni.usc.edu/wp-content/uploads/how_to_apply/ADNI_Acknowledgement_List.pdf
| | - Caitlin A. Finney
- Neuroinflammation Research Group, Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, Sydney, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
9
|
Chaloemtoem A, Thornton V, Chang Y, Anokhin AP, Belloy ME, Bijsterbosch J, Gordon BA, Hartz SM, Bierut LJ. Hippocampal volumes in UK Biobank are associated with APOE only in older adults. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.05.24307704. [PMID: 38883747 PMCID: PMC11177915 DOI: 10.1101/2024.06.05.24307704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
INTRODUCTION The hippocampus atrophies with age and is implicated in neurodegenerative disorders including Alzheimer's disease (AD). We examined the interplay between age and APOE genotype on total hippocampal volume. METHODS Utilizing neuroimaging data from 37,463 UK Biobank participants, we applied linear regression to quantify the association of age and APOE with hippocampal volume and identified the age when volumes of ε2/ε3, ε3/ε4, and ε4/ε4 carriers significantly deviated from ε3/ε3 using generalized additive modeling. RESULTS Total hippocampal volume declined with age, with significant differences by APOE genotype emerging after age 60. ε3/ε4 and ε4/ε4 carriers displayed reduced volumes from ages 69 and 61, respectively, while ε2/ε3 showed delayed decline starting at age 76. DISCUSSION The association of APOE and hippocampal volume is age-dependent, with differences in volumes of ε4/ε4 carriers detected as early as age 61. This work underscores the importance of APOE genotype in determining when to begin screening for AD.
Collapse
|
10
|
Bašić J, Milošević V, Djordjević B, Stojiljković V, Živanović M, Stefanović N, Aracki Trenkić A, Stojanov D, Jevtović Stoimenov T, Stojanović I. Matrix Remodeling Enzymes as Potential Fluid Biomarkers of Neurodegeneration in Alzheimer's Disease. Int J Mol Sci 2024; 25:5703. [PMID: 38891891 PMCID: PMC11171655 DOI: 10.3390/ijms25115703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/17/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
This study investigated the diagnostic accuracy of plasma biomarkers-specifically, matrix metalloproteinase (MMP-9), tissue inhibitor of metalloproteinase (TIMP-1), CD147, and the MMP-/TIMP-1 ratio in patients with Alzheimer's disease (AD) dementia. The research cohort comprised patients diagnosed with probable AD dementia and a control group of cognitively unimpaired (CU) individuals. Neuroradiological assessments included brain magnetic resonance imaging (MRI) following dementia protocols, with subsequent volumetric analysis. Additionally, cerebrospinal fluid (CSF) AD biomarkers were classified using the A/T/N system, and apolipoprotein E (APOE) ε4 carrier status was determined. Findings revealed elevated plasma levels of MMP-9 and TIMP-1 in AD dementia patients compared to CU individuals. Receiver operating characteristic (ROC) curve analysis demonstrated significant differences in the areas under the curve (AUC) for MMP-9 (p < 0.001) and TIMP-1 (p < 0.001). Notably, plasma TIMP-1 levels were significantly lower in APOE ε4+ patients than in APOE ε4- patients (p = 0.041). Furthermore, APOE ε4+ patients exhibited reduced hippocampal volume, particularly in total, right, and left hippocampal measurements. TIMP-1 levels exhibited a positive correlation, while the MMP-9/TIMP-1 ratio showed a negative correlation with hippocampal volume parameters. This study sheds light on the potential use of TIMP-1 as a diagnostic marker and its association with hippocampal changes in AD.
Collapse
Affiliation(s)
- Jelena Bašić
- Department of Biochemistry, Faculty of Medicine, University of Niš, 18000 Niš, Serbia; (B.D.); (V.S.); (T.J.S.); (I.S.)
| | - Vuk Milošević
- Faculty of Medicine, University of Niš, 18000 Niš, Serbia; (V.M.); (A.A.T.); (D.S.)
- Clinic of Neurology, University Clinical Center Niš, 18000 Niš, Serbia
| | - Branka Djordjević
- Department of Biochemistry, Faculty of Medicine, University of Niš, 18000 Niš, Serbia; (B.D.); (V.S.); (T.J.S.); (I.S.)
| | - Vladana Stojiljković
- Department of Biochemistry, Faculty of Medicine, University of Niš, 18000 Niš, Serbia; (B.D.); (V.S.); (T.J.S.); (I.S.)
| | - Milica Živanović
- Center for Radiology, University Clinical Center Niš, 18000 Niš, Serbia;
| | - Nikola Stefanović
- Department of Pharmacy, Faculty of Medicine, University of Niš, 18000 Niš, Serbia;
| | - Aleksandra Aracki Trenkić
- Faculty of Medicine, University of Niš, 18000 Niš, Serbia; (V.M.); (A.A.T.); (D.S.)
- Center for Radiology, University Clinical Center Niš, 18000 Niš, Serbia;
| | - Dragan Stojanov
- Faculty of Medicine, University of Niš, 18000 Niš, Serbia; (V.M.); (A.A.T.); (D.S.)
- Center for Radiology, University Clinical Center Niš, 18000 Niš, Serbia;
| | - Tatjana Jevtović Stoimenov
- Department of Biochemistry, Faculty of Medicine, University of Niš, 18000 Niš, Serbia; (B.D.); (V.S.); (T.J.S.); (I.S.)
| | - Ivana Stojanović
- Department of Biochemistry, Faculty of Medicine, University of Niš, 18000 Niš, Serbia; (B.D.); (V.S.); (T.J.S.); (I.S.)
| |
Collapse
|
11
|
Sakurai R, Pieruccini‐Faria F, Cornish B, Fraser J, Binns MA, Beaton D, Dilliott AA, Kwan D, Ramirez J, Tan B, Scott CJM, Sunderland KM, Tartaglia C, Finger E, Zinman L, Freedman M, McLaughlin PM, Swartz RH, Symons S, Lang AE, Bartha R, Black SE, Masellis M, Hegele RA, McIlroy W, Montero‐Odasso M. Link among apolipoprotein E E4, gait, and cognition in neurodegenerative diseases: ONDRI study. Alzheimers Dement 2024; 20:2968-2979. [PMID: 38470007 PMCID: PMC11032526 DOI: 10.1002/alz.13740] [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: 06/16/2023] [Revised: 12/08/2023] [Accepted: 12/14/2023] [Indexed: 03/13/2024]
Abstract
INTRODUCTION Apolipoprotein E E4 allele (APOE E4) and slow gait are independently associated with cognitive impairment and dementia. However, it is unknown whether their coexistence is associated with poorer cognitive performance and its underlying mechanism in neurodegenerative diseases. METHODS Gait speed, APOE E4, cognition, and neuroimaging were assessed in 480 older adults with neurodegeneration. Participants were grouped by APOE E4 presence and slow gait. Mediation analyses were conducted to determine if brain structures could explain the link between these factors and cognitive performance. RESULTS APOE E4 carriers with slow gait had the lowest global cognitive performance and smaller gray matter volumes compared to non-APOE E4 carriers with normal gait. Coexistence of APOE E4 and slow gait best predicted global and domain-specific poorer cognitive performances, mediated by smaller gray matter volume. DISCUSSION Gait slowness in APOE E4 carriers with neurodegenerative diseases may indicate extensive gray matter changes associated with poor cognition. HIGHLIGHTS APOE E4 and slow gait are risk factors for cognitive decline in neurodegenerative diseases. Slow gait and smaller gray matter volumes are associated, independently of APOE E4. Worse cognition in APOE E4 carriers with slow gait is explained by smaller GM volume. Gait slowness in APOE E4 carriers indicates poorer cognition-related brain changes.
Collapse
Affiliation(s)
- Ryota Sakurai
- Research Team for Social Participation and Healthy AgingTokyo Metropolitan Institute for Geriatrics and GerontologyItabashi‐kuTokyoJapan
- Gait & Brain Lab, St. Joseph' Health Care London, Lawson Health Research, Western University, Division of Geriatric MedicineLondonOntarioCanada
| | - Frederico Pieruccini‐Faria
- Gait & Brain Lab, St. Joseph' Health Care London, Lawson Health Research, Western University, Division of Geriatric MedicineLondonOntarioCanada
- Department of MedicineDivision of Geriatric MedicineParkwood HospitalWestern University, Parkwood InstituteLondonOntarioCanada
| | - Benjamin Cornish
- Neuroscience, Mobility and Balance Lab, Department of Kinesiology and Health SciencesUniversity of WaterlooWaterlooOntarioCanada
| | - Julia Fraser
- Neuroscience, Mobility and Balance Lab, Department of Kinesiology and Health SciencesUniversity of WaterlooWaterlooOntarioCanada
| | - Malcolm A. Binns
- Rotman Research Institute, Baycrest Health SciencesTorontoOntarioCanada
| | - Derek Beaton
- Data Science and Advanced Analytics, St. Michael's Hospital, Unity Health TorontoTorontoOntarioCanada
| | - Allison Ann Dilliott
- Department of Neurology and NeurosurgeryMontreal Neurological Institute, McGill UniversityMontréalQuebecCanada
| | - Donna Kwan
- Centre for Neuroscience Studies, Queen's UniversityKingstonOntarioCanada
| | - Joel Ramirez
- L.C. Campbell Cognitive Neurology Research Unit, Hurvitz Brain Sciences Program, Department of Medicine (Neurology)Sunnybrook Research Institute, Sunnybrook HSC, University of TorontoTorontoOntarioCanada
| | - Brian Tan
- Rotman Research Institute, Baycrest Health SciencesTorontoOntarioCanada
| | | | | | - Carmela Tartaglia
- Krembil Brain InstituteUniversity Health Network Memory Clinic, Toronto Western HospitalTorontoOntarioCanada
- Tanz Centre for Research in Neurodegenerative Diseases, University of TorontoTorontoOntarioCanada
| | - Elizabeth Finger
- Department of Clinical Neurological SciencesSchulich School of Medicine and Dentistry, Western UniversityLondonOntarioCanada
| | - Lorne Zinman
- Sunnybrook Research Institute, Sunnybrook Health Sciences CentreTorontoOntarioCanada
- Department of Medicine (Neurology)University of TorontoTorontoOntarioCanada
| | - Morris Freedman
- Rotman Research Institute, Baycrest Health SciencesTorontoOntarioCanada
- Department of Medicine (Neurology)University of TorontoTorontoOntarioCanada
- Division of NeurologyBaycrest Health SciencesTorontoOntarioCanada
| | - Paula M. McLaughlin
- Halifax Clinical Psychology Residency ProgramNova Scotia Health AuthorityHalifaxNova ScotiaCanada
| | - Richard H. Swartz
- Sunnybrook Research Institute, Sunnybrook Health Sciences CentreTorontoOntarioCanada
- Department of Medicine (Neurology)University of TorontoTorontoOntarioCanada
| | - Sean Symons
- L.C. Campbell Cognitive Neurology Research Unit, Hurvitz Brain Sciences Program, Department of Medicine (Neurology)Sunnybrook Research Institute, Sunnybrook HSC, University of TorontoTorontoOntarioCanada
| | - Anthony E. Lang
- Division of NeurologyDepartment of MedicineEdmond J Safra Program in Parkinson's Disease and Morton and Gloria Shulman Movement Disorders ClinicToronto Western HospitalUniversity of TorontoTorontoOntarioCanada
| | - Robert Bartha
- Department of Medical BiophysicsSchulich School of Medicine and Dentistry, Robarts Research Institute, Western UniversityLondonOntarioCanada
| | - Sandra E. Black
- L.C. Campbell Cognitive Neurology Research Unit, Hurvitz Brain Sciences Program, Department of Medicine (Neurology)Sunnybrook Research Institute, Sunnybrook HSC, University of TorontoTorontoOntarioCanada
| | - Mario Masellis
- L.C. Campbell Cognitive Neurology Research Unit, Hurvitz Brain Sciences Program, Department of Medicine (Neurology)Sunnybrook Research Institute, Sunnybrook HSC, University of TorontoTorontoOntarioCanada
| | - Robert A. Hegele
- Schulich School of Medicine and Dentistry, Western UniversityLondonOntarioCanada
- Robarts Research Institute, Western UniversityLondonOntarioCanada
| | - William McIlroy
- Neuroscience, Mobility and Balance Laboratory, Department of Kinesiology and Health SciencesUniversity of WaterlooWaterlooOntarioCanada
| | - ONDRI Investigators
- Research Team for Social Participation and Healthy AgingTokyo Metropolitan Institute for Geriatrics and GerontologyItabashi‐kuTokyoJapan
| | - Manuel Montero‐Odasso
- Gait & Brain Lab, St. Joseph' Health Care London, Lawson Health Research, Western University, Division of Geriatric MedicineLondonOntarioCanada
- Gait and Brain Lab, Division of Geriatric Medicineand Lawson Health Research InstituteParkwood Institute, Western UniversityLondonOntarioCanada
- Division of Geriatric MedicineDepartment of MedicineSchulich School of Medicine and Dentistry, Western University, Parkwood InstituteLondonOntarioCanada
| |
Collapse
|
12
|
Onos K, Lin PB, Pandey RS, Persohn SA, Burton CP, Miner EW, Eldridge K, Kanyinda JN, Foley KE, Carter GW, Howell GR, Territo PR. Assessment of Neurovascular Uncoupling: APOE Status is a Key Driver of Early Metabolic and Vascular Dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.13.571584. [PMID: 38168292 PMCID: PMC10760108 DOI: 10.1101/2023.12.13.571584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common cause of dementia worldwide, with apolipoprotein ε4 (APOEε4) being the strongest genetic risk factor. Current clinical diagnostic imaging focuses on amyloid and tau; however, new methods are needed for earlier detection. METHODS PET imaging was used to assess metabolism-perfusion in both sexes of aging C57BL/6J, and hAPOE mice, and were verified by transcriptomics, and immunopathology. RESULTS All hAPOE strains showed AD phenotype progression by 8 mo, with females exhibiting the regional changes, which correlated with GO-term enrichments for glucose metabolism, perfusion, and immunity. Uncoupling analysis revealed APOEε4/ε4 exhibited significant Type-1 uncoupling (↓ glucose uptake, ↑ perfusion) at 8 and 12 mo, while APOEε3/ε4 demonstrated Type-2 uncoupling (↑ glucose uptake, ↓ perfusion), while immunopathology confirmed cell specific contributions. DISCUSSION This work highlights APOEε4 status in AD progression manifest as neurovascular uncoupling driven by immunological activation, and may serve as an early diagnostic biomarker.
Collapse
Affiliation(s)
- Kristen Onos
- The Jackson Laboratory, Bar Harbor, ME 04609 USA
| | - Peter B. Lin
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202 USA
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Ravi S. Pandey
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032 USA
| | - Scott A. Persohn
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Charles P. Burton
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Ethan W. Miner
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Kierra Eldridge
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | | | - Kate E. Foley
- The Jackson Laboratory, Bar Harbor, ME 04609 USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Gregory W. Carter
- The Jackson Laboratory, Bar Harbor, ME 04609 USA
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032 USA
| | | | - Paul R. Territo
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202 USA
- Department of Medicine, Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis IN 46202 USA
| |
Collapse
|
13
|
Jung M, Jung JS, Pfeifer J, Hartmann C, Ehrhardt T, Abid CL, Kintzel J, Puls A, Navarrete Santos A, Hollemann T, Riemann D, Rujescu D. Neuronal Stem Cells from Late-Onset Alzheimer Patients Show Altered Regulation of Sirtuin 1 Depending on Apolipoprotein E Indicating Disturbed Stem Cell Plasticity. Mol Neurobiol 2024; 61:1562-1579. [PMID: 37728850 PMCID: PMC10896791 DOI: 10.1007/s12035-023-03633-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 08/31/2023] [Indexed: 09/21/2023]
Abstract
Late-onset Alzheimer's disease (AD) is a complex multifactorial disease. The greatest known risk factor for late-onset AD is the E4 allele of the apolipoprotein E (APOE), while increasing age is the greatest known non-genetic risk factor. The cell type-specific functions of neural stem cells (NSCs), in particular their stem cell plasticity, remain poorly explored in the context of AD pathology. Here, we describe a new model that employs late-onset AD patient-derived induced pluripotent stem cells (iPSCs) to generate NSCs and to examine the role played by APOE4 in the expression of aging markers such as sirtuin 1 (SIRT1) in comparison to healthy subjects carrying APOE3. The effect of aging was investigated by using iPSC-derived NSCs from old age subjects as healthy matched controls. Transcript and protein analysis revealed that genes were expressed differently in NSCs from late-onset AD patients, e.g., exhibiting reduced autophagy-related protein 7 (ATG7), phosphatase and tensin homolog (PTEN), and fibroblast growth factor 2 (FGF2). Since SIRT1 expression differed between APOE3 and APOE4 NSCs, the suppression of APOE function in NSCs also repressed the expression of SIRT1. However, the forced expression of APOE3 by plasmids did not recover differently expressed genes. The altered aging markers indicate decreased plasticity of NSCs. Our study provides a suitable in vitro model to investigate changes in human NSCs associated with aging, APOE4, and late-onset AD.
Collapse
Affiliation(s)
- Matthias Jung
- Institute of Physiological Chemistry (IPC), Faculty of Medicine, Martin Luther University Halle-Wittenberg, Hollystrasse 1, 06114, Halle (Saale), Germany.
| | - Juliane-Susanne Jung
- Institute of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06118, Halle (Saale), Germany
| | - Jenny Pfeifer
- Institute of Physiological Chemistry (IPC), Faculty of Medicine, Martin Luther University Halle-Wittenberg, Hollystrasse 1, 06114, Halle (Saale), Germany
| | - Carla Hartmann
- Institute of Physiological Chemistry (IPC), Faculty of Medicine, Martin Luther University Halle-Wittenberg, Hollystrasse 1, 06114, Halle (Saale), Germany
| | - Toni Ehrhardt
- Institute of Physiological Chemistry (IPC), Faculty of Medicine, Martin Luther University Halle-Wittenberg, Hollystrasse 1, 06114, Halle (Saale), Germany
| | - Chaudhry Luqman Abid
- Institute of Physiological Chemistry (IPC), Faculty of Medicine, Martin Luther University Halle-Wittenberg, Hollystrasse 1, 06114, Halle (Saale), Germany
| | - Jenny Kintzel
- Institute of Physiological Chemistry (IPC), Faculty of Medicine, Martin Luther University Halle-Wittenberg, Hollystrasse 1, 06114, Halle (Saale), Germany
| | - Anne Puls
- Institute of Physiological Chemistry (IPC), Faculty of Medicine, Martin Luther University Halle-Wittenberg, Hollystrasse 1, 06114, Halle (Saale), Germany
| | - Anne Navarrete Santos
- Institute of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06118, Halle (Saale), Germany
| | - Thomas Hollemann
- Institute of Physiological Chemistry (IPC), Faculty of Medicine, Martin Luther University Halle-Wittenberg, Hollystrasse 1, 06114, Halle (Saale), Germany
| | - Dagmar Riemann
- Department Medical Immunology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Magdeburger Strasse 2, 06112, Halle (Saale), Germany
| | - Dan Rujescu
- Department of Psychiatry and Psychotherapy, Division of General Psychiatry, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| |
Collapse
|
14
|
Garcia P, Mendoza L, Padron D, Duarte A, Duara R, Loewenstein D, Greig-Custo M, Barker W, Curiel R, Rosselli M, Rodriguez M. Sex significantly predicts medial temporal volume when controlling for the influence of ApoE4 biomarker and demographic variables: A cross-ethnic comparison. J Int Neuropsychol Soc 2024; 30:128-137. [PMID: 37385978 PMCID: PMC11057967 DOI: 10.1017/s1355617723000358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
OBJECTIVE To explore the relationship between age, education, sex, and ApoE4 (+) status to brain volume among a cohort with amnestic mild cognitive impairment (aMCI). METHOD One hundred and twenty-three participants were stratified into Hispanic (n = 75) and White non-Hispanic (WNH, N = 48). Multiple linear regression analyses were conducted with age, education, sex, and ApoE4 status as predictor variables and left and right combined MRI volumes of the hippocampus, parahippocampus, and entorhinal cortex as dependent variables. Variations in head sizes were corrected by normalization with a total intracranial volume measurement. RESULTS Bonferroni-corrected results indicated that when controlling for ApoE4 status, education, and age, sex was a significant predictor of hippocampal volume among the Hispanic group (β = .000464, R2 = .196, p < .01) and the WNH group (β = .000455, R2 = .195, p < .05). Education (β = .000028, R2 = .168, p < .01) and sex (β = .000261, R2 = .168, p < .01) were significant predictors of parahippocampal volume among the Hispanic MCI group when controlling for the effects of ApoE4 status and age. One-way ANCOVAs comparing hippocampal and parahippocampal volume between males and females within groups revealed that females had significantly larger hippocampal volumes (p < .05). Hispanic females had significantly larger hippocampal (p < .001) and parahippocampal (p < .05) volume compared to males. No sex differences in parahippocampal volume were noted among WNHs. CONCLUSIONS Biological sex, rather than ApoE4 status, was a greater predictor of hippocampal volume among Hispanic and WNH females. These findings add to the mixed literature on sex differences in dementia research and highlight continued emphasis on ethnic populations to elucidate on neurodegenerative disparities.
Collapse
Affiliation(s)
- Patricia Garcia
- Department of Clinical Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | | | - Ranjan Duara
- Mount Sinai Medical Center, Miami Beach, FL, USA
| | - David Loewenstein
- University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | | | | | - Rosie Curiel
- University of Miami Leonard M. Miller School of Medicine, Miami, FL, USA
| | - Monica Rosselli
- Department of Psychology, Florida Atlantic University, Davie, FL, USA
| | - Miriam Rodriguez
- Department of Health & Wellness Design, Indiana University Bloomington School of Public Health, Bloomington, IN, USA
| |
Collapse
|
15
|
Ji X, Peng X, Tang H, Pan H, Wang W, Wu J, Chen J, Wei N. Alzheimer's disease phenotype based upon the carrier status of the apolipoprotein E ɛ4 allele. Brain Pathol 2024; 34:e13208. [PMID: 37646624 PMCID: PMC10711266 DOI: 10.1111/bpa.13208] [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: 12/06/2022] [Accepted: 08/05/2023] [Indexed: 09/01/2023] Open
Abstract
The apolipoprotein E ɛ4 allele (APOE4) is universally acknowledged as the most potent genetic risk factor for Alzheimer's disease (AD). APOE4 promotes the initiation and progression of AD. Although the underlying mechanisms are unclearly understood, differences in lipid-bound affinity among the three APOE isoforms may constitute the basis. The protein APOE4 isoform has a high affinity with triglycerides and cholesterol. A distinction in lipid metabolism extensively impacts neurons, microglia, and astrocytes. APOE4 carriers exhibit phenotypic differences from non-carriers in clinical examinations and respond differently to multiple treatments. Therefore, we hypothesized that phenotypic classification of AD patients according to the status of APOE4 carrier will help specify research and promote its use in diagnosing and treating AD. Recent reviews have mainly evaluated the differences between APOE4 allele carriers and non-carriers from gene to protein structures, clinical features, neuroimaging, pathology, the neural network, and the response to various treatments, and have provided the feasibility of phenotypic group classification based on APOE4 carrier status. This review will facilitate the application of APOE phenomics concept in clinical practice and promote further medical research on AD.
Collapse
Affiliation(s)
- Xiao‐Yu Ji
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeGuangdongChina
- Brain Function and Disease LaboratoryShantou University Medical CollegeGuangdongChina
| | - Xin‐Yuan Peng
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeGuangdongChina
| | - Hai‐Liang Tang
- Fudan University Huashan Hospital, Department of Neurosurgery, State Key Laboratory for Medical NeurobiologyInstitutes of Brain Science, Shanghai Medical College‐Fudan UniversityShanghaiChina
| | - Hui Pan
- Shantou Longhu People's HospitalShantouGuangdongChina
| | - Wei‐Tang Wang
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeGuangdongChina
| | - Jie Wu
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeGuangdongChina
- Brain Function and Disease LaboratoryShantou University Medical CollegeGuangdongChina
| | - Jian Chen
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeGuangdongChina
| | - Nai‐Li Wei
- Department of NeurosurgeryThe First Affiliated Hospital of Shantou University Medical CollegeGuangdongChina
| |
Collapse
|
16
|
Wang X, Zhu J, Pan W, Zhu J, Zhang H. Nonparametric Statistical Inference via Metric Distribution Function in Metric Spaces. J Am Stat Assoc 2023; 119:2772-2784. [PMID: 40078669 PMCID: PMC11902916 DOI: 10.1080/01621459.2023.2277417] [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: 07/13/2021] [Revised: 10/11/2023] [Accepted: 10/18/2023] [Indexed: 03/14/2025]
Abstract
The distribution function is essential in statistical inference and connected with samples to form a directed closed loop by the correspondence theorem in measure theory and the Glivenko-Cantelli and Donsker properties. This connection creates a paradigm for statistical inference. However, existing distribution functions are defined in Euclidean spaces and are no longer convenient to use in rapidly evolving data objects of complex nature. It is imperative to develop the concept of the distribution function in a more general space to meet emerging needs. Note that the linearity allows us to use hypercubes to define the distribution function in a Euclidean space. Still, without the linearity in a metric space, we must work with the metric to investigate the probability measure. We introduce a class of metric distribution functions through the metric only. We overcome this challenging step by proving the correspondence theorem and the Glivenko-Cantelli theorem for metric distribution functions in metric spaces, laying the foundation for conducting rational statistical inference for metric space-valued data. Then, we develop a homogeneity test and a mutual independence test for non-Euclidean random objects and present comprehensive empirical evidence to support the performance of our proposed methods.
Collapse
Affiliation(s)
- Xueqin Wang
- University of Science and Technology of China, Sun Yat-Sen University
- London School of Economics and Political Science, Chinese Academy of Sciences Yale University
| | - Jin Zhu
- University of Science and Technology of China, Sun Yat-Sen University
- London School of Economics and Political Science, Chinese Academy of Sciences Yale University
| | - Wenliang Pan
- University of Science and Technology of China, Sun Yat-Sen University
- London School of Economics and Political Science, Chinese Academy of Sciences Yale University
| | - Junhao Zhu
- University of Science and Technology of China, Sun Yat-Sen University
- London School of Economics and Political Science, Chinese Academy of Sciences Yale University
| | - Heping Zhang
- University of Science and Technology of China, Sun Yat-Sen University
- London School of Economics and Political Science, Chinese Academy of Sciences Yale University
| |
Collapse
|
17
|
Christopher-Hayes NJ, Embury CM, Wiesman AI, May PE, Schantell M, Johnson CM, Wolfson SL, Murman DL, Wilson TW. Piecing it together: atrophy profiles of hippocampal subfields relate to cognitive impairment along the Alzheimer's disease spectrum. Front Aging Neurosci 2023; 15:1212197. [PMID: 38020776 PMCID: PMC10644116 DOI: 10.3389/fnagi.2023.1212197] [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: 04/25/2023] [Accepted: 10/13/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction People with Alzheimer's disease (AD) experience more rapid declines in their ability to form hippocampal-dependent memories than cognitively normal healthy adults. Degeneration of the whole hippocampal formation has previously been found to covary with declines in learning and memory, but the associations between subfield-specific hippocampal neurodegeneration and cognitive impairments are not well characterized in AD. To improve prognostic procedures, it is critical to establish in which hippocampal subfields atrophy relates to domain-specific cognitive declines among people along the AD spectrum. In this study, we examine high-resolution structural magnetic resonance imaging (MRI) of the medial temporal lobe and extensive neuropsychological data from 29 amyloid-positive people on the AD spectrum and 17 demographically-matched amyloid-negative healthy controls. Methods Participants completed a battery of neuropsychological exams including select tests of immediate recollection, delayed recollection, and general cognitive status (i.e., performance on the Mini-Mental State Examination [MMSE] and Montreal Cognitive Assessment [MoCA]). Hippocampal subfield volumes (CA1, CA2, CA3, dentate gyrus, and subiculum) were measured using a dedicated MRI slab sequence targeting the medial temporal lobe and used to compute distance metrics to quantify AD spectrum-specific atrophic patterns and their impact on cognitive outcomes. Results Our results replicate prior studies showing that CA1, dentate gyrus, and subiculum hippocampal subfield volumes were significantly reduced in AD spectrum participants compared to amyloid-negative controls, whereas CA2 and CA3 did not exhibit such patterns of atrophy. Moreover, degeneration of the subiculum along the AD spectrum was linked to a significant decline in general cognitive status measured by the MMSE, while degeneration scores of the CA1 and dentate gyrus were more widely associated with declines on the MMSE and tests of learning and memory. Discussion These findings provide evidence that subfield-specific patterns of hippocampal degeneration, in combination with cognitive assessments, may constitute a sensitive prognostic approach and could be used to better track disease trajectories among individuals on the AD spectrum.
Collapse
Affiliation(s)
- Nicholas J. Christopher-Hayes
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, United States
- Center for Mind and Brain, University of California, Davis, CA, United States
| | - Christine M. Embury
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, United States
- Department of Psychology, University of Nebraska at Omaha, Omaha, NE, United States
| | - Alex I. Wiesman
- Montreal Neurological Institute, McGill University, Montréal, QC, Canada
| | - Pamela E. May
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, United States
| | - Mikki Schantell
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, United States
- College of Medicine, UNMC, Omaha, NE, United States
| | | | | | - Daniel L. Murman
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, United States
- Memory Disorders and Behavioral Neurology Program, UNMC, Omaha, NE, United States
| | - Tony W. Wilson
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE, United States
- College of Medicine, UNMC, Omaha, NE, United States
- Department of Pharmacology and Neuroscience, Creighton University, Omaha, NE, United States
| |
Collapse
|
18
|
Huang W, Zeng J, Jia L, Zhu D, O’Brien J, Ritchie C, Shu N, Su L. Genetic risks of Alzheimer's by APOE and MAPT on cortical morphology in young healthy adults. Brain Commun 2023; 5:fcad234. [PMID: 37693814 PMCID: PMC10489122 DOI: 10.1093/braincomms/fcad234] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 07/29/2023] [Accepted: 08/30/2023] [Indexed: 09/12/2023] Open
Abstract
Genetic risk factors such as APOE ε4 and MAPT (rs242557) A allele are associated with amyloid and tau pathways and grey matter changes at both early and established stages of Alzheimer's disease, but their effects on cortical morphology in young healthy adults remain unclear. A total of 144 participants aged from 18 to 24 underwent 3T MRI and genotyping for APOE and MAPT to investigate unique impacts of these genetic risk factors in a cohort without significant comorbid conditions such as metabolic and cardiovascular diseases. We segmented the cerebral cortex into 68 regions and calculated the cortical area, thickness, curvature and folding index for each region. Then, we trained machine learning models to classify APOE and MAPT genotypes using these morphological features. In addition, we applied a growing hierarchical self-organizing maps algorithm, which clustered the 68 regions into 4 subgroups representing different morphological patterns. Then, we performed general linear model analyses to estimate the interaction between APOE and MAPT on cortical patterns. We found that the classifiers using all cortical features could accurately classify individuals carrying genetic risks of dementia outperforming each individual feature alone. APOE ε4 carriers had a more convoluted and thinner cortex across the cerebral cortex. A similar pattern was found in MAPT A allele carriers only in the regions that are vulnerable for early tau pathology. With the clustering analysis, we found a synergetic effect between APOE ε4 and MAPT A allele, i.e. carriers of both risk factors showed the most deviation of cortical pattern from the typical pattern of that cluster. Genetic risk factors of dementia by APOE ε4 and MAPT (rs242557) A allele were associated with variations of cortical morphology, which can be observed in young healthy adults more than 30 years before Alzheimer's pathology is likely to occur and 50 years before dementia symptoms may begin.
Collapse
Affiliation(s)
- Weijie Huang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
- Department of Neuroscience, Neuroscience Institute, Insigneo Institute for In Silico Medicine, University of Sheffield, Sheffield S10 2HQ, UK
- School of Systems Science, Beijing Normal University, Beijing 100875, China
| | - Jianmin Zeng
- Faculty of Psychology, Sino-Britain Centre for Cognition and Ageing Research, Southwest University, Chongqing 400715, China
| | - Lina Jia
- Beijing Anding Hospital, Capital Medical University, Beijing 100088, China
| | - Dajiang Zhu
- Department of Computer Science and Engineering, University of Texas at Arlington, Arlington, TX 76019, USA
| | - John O’Brien
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Craig Ritchie
- Edinburgh Dementia Prevention and Centre for Clinical Brain Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh EH4 2XU, UK
- Scottish Brain Sciences, Edinburgh EH12 9DQ, UK
| | - Ni Shu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing 100875, China
| | - Li Su
- Department of Neuroscience, Neuroscience Institute, Insigneo Institute for In Silico Medicine, University of Sheffield, Sheffield S10 2HQ, UK
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge CB2 0SZ, UK
| |
Collapse
|
19
|
Calderón-Garcidueñas L, Hernández-Luna J, Aiello-Mora M, Brito-Aguilar R, Evelson PA, Villarreal-Ríos R, Torres-Jardón R, Ayala A, Mukherjee PS. APOE Peripheral and Brain Impact: APOE4 Carriers Accelerate Their Alzheimer Continuum and Have a High Risk of Suicide in PM 2.5 Polluted Cities. Biomolecules 2023; 13:927. [PMID: 37371506 DOI: 10.3390/biom13060927] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/22/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
This Review emphasizes the impact of APOE4-the most significant genetic risk factor for Alzheimer's disease (AD)-on peripheral and neural effects starting in childhood. We discuss major mechanistic players associated with the APOE alleles' effects in humans to understand their impact from conception through all life stages and the importance of detrimental, synergistic environmental exposures. APOE4 influences AD pathogenesis, and exposure to fine particulate matter (PM2.5), manufactured nanoparticles (NPs), and ultrafine particles (UFPs) associated with combustion and friction processes appear to be major contributors to cerebrovascular dysfunction, neuroinflammation, and oxidative stress. In the context of outdoor and indoor PM pollution burden-as well as Fe, Ti, and Al alloys; Hg, Cu, Ca, Sn, and Si UFPs/NPs-in placenta and fetal brain tissues, urban APOE3 and APOE4 carriers are developing AD biological disease hallmarks (hyperphosphorylated-tau (P-tau) and amyloid beta 42 plaques (Aβ42)). Strikingly, for Metropolitan Mexico City (MMC) young residents ≤ 40 y, APOE4 carriers have 4.92 times higher suicide odds and 23.6 times higher odds of reaching Braak NFT V stage versus APOE4 non-carriers. The National Institute on Aging and Alzheimer's Association (NIA-AA) framework could serve to test the hypothesis that UFPs and NPs are key players for oxidative stress, neuroinflammation, protein aggregation and misfolding, faulty complex protein quality control, and early damage to cell membranes and organelles of neural and vascular cells. Noninvasive biomarkers indicative of the P-tau and Aβ42 abnormal protein deposits are needed across the disease continuum starting in childhood. Among the 21.8 million MMC residents, we have potentially 4 million APOE4 carriers at accelerated AD progression. These APOE4 individuals are prime candidates for early neuroprotective interventional trials. APOE4 is key in the development of AD evolving from childhood in highly polluted urban centers dominated by anthropogenic and industrial sources of pollution. APOE4 subjects are at higher early risk of AD development, and neuroprotection ought to be implemented. Effective reductions of PM2.5, UFP, and NP emissions from all sources are urgently needed. Alzheimer's Disease prevention ought to be at the core of the public health response and physicians-scientist minority research be supported.
Collapse
Affiliation(s)
- Lilian Calderón-Garcidueñas
- College of Health, The University of Montana, Missoula, MT 59812, USA
- Universidad del Valle de México, Mexico City 14370, Mexico
| | | | - Mario Aiello-Mora
- Otorrinolaryngology Department, Instituto Nacional de Cardiología, Mexico City 14080, Mexico
| | | | - Pablo A Evelson
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires C1113 AAD, Argentina
| | | | - Ricardo Torres-Jardón
- Instituto de Ciencias de la Atmósfera y Cambio Climático, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Alberto Ayala
- Sacramento Metropolitan Air Quality Management District, Sacramento, CA 95814, USA
- West Virginia University, Morgantown, WV 26506, USA
| | - Partha S Mukherjee
- Interdisciplinary Statistical Research Unit, Indian Statistical Institute, Kolkata 700108, India
| |
Collapse
|
20
|
Gorelik AJ, Paul SE, Karcher NR, Johnson EC, Nagella I, Blaydon L, Modi H, Hansen IS, Colbert SMC, Baranger DAA, Norton SA, Spears I, Gordon B, Zhang W, Hill PL, Oltmanns TF, Bijsterbosch JD, Agrawal A, Hatoum AS, Bogdan R. A Phenome-Wide Association Study (PheWAS) of Late Onset Alzheimer Disease Genetic Risk in Children of European Ancestry at Middle Childhood: Results from the ABCD Study. Behav Genet 2023; 53:249-264. [PMID: 37071275 PMCID: PMC10309061 DOI: 10.1007/s10519-023-10140-3] [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: 11/19/2022] [Accepted: 03/08/2023] [Indexed: 04/19/2023]
Abstract
Genetic risk for Late Onset Alzheimer Disease (AD) has been associated with lower cognition and smaller hippocampal volume in healthy young adults. However, whether these and other associations are present during childhood remains unclear. Using data from 5556 genomically-confirmed European ancestry youth who completed the baseline session of the ongoing the Adolescent Brain Cognitive DevelopmentSM Study (ABCD Study®), our phenome-wide association study estimating associations between four indices of genetic risk for late-onset AD (i.e., AD polygenic risk scores (PRS), APOE rs429358 genotype, AD PRS with the APOE region removed (ADPRS-APOE), and an interaction between ADPRS-APOE and APOE genotype) and 1687 psychosocial, behavioral, and neural phenotypes revealed no significant associations after correction for multiple testing (all ps > 0.0002; all pfdr > 0.07). These data suggest that AD genetic risk may not phenotypically manifest during middle-childhood or that effects are smaller than this sample is powered to detect.
Collapse
Affiliation(s)
- Aaron J Gorelik
- Department of Psychological and Brain Sciences, Washington University in Saint Louis, One Booking Drive, St. Louis, MO, 63130, USA
| | - Sarah E Paul
- Department of Psychological and Brain Sciences, Washington University in Saint Louis, One Booking Drive, St. Louis, MO, 63130, USA
| | - Nicole R Karcher
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Emma C Johnson
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Isha Nagella
- Department of Psychological and Brain Sciences, Washington University in Saint Louis, One Booking Drive, St. Louis, MO, 63130, USA
| | - Lauren Blaydon
- Department of Psychological and Brain Sciences, Washington University in Saint Louis, One Booking Drive, St. Louis, MO, 63130, USA
| | - Hailey Modi
- Department of Psychological and Brain Sciences, Washington University in Saint Louis, One Booking Drive, St. Louis, MO, 63130, USA
| | - Isabella S Hansen
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Sarah M C Colbert
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - David A A Baranger
- Department of Psychological and Brain Sciences, Washington University in Saint Louis, One Booking Drive, St. Louis, MO, 63130, USA
| | - Sara A Norton
- Department of Psychological and Brain Sciences, Washington University in Saint Louis, One Booking Drive, St. Louis, MO, 63130, USA
| | - Isaiah Spears
- Department of Psychological and Brain Sciences, Washington University in Saint Louis, One Booking Drive, St. Louis, MO, 63130, USA
| | - Brian Gordon
- Department of Radiology, Washington University in Saint Louis, 660 South Euclid Ave, Box 8225, St. Louis, MO, 63110, USA
- Charles F. and Joanne Knight Alzheimer Disease Research Center, Washington University, St Louis, MO, USA
| | - Wei Zhang
- Department of Radiology, Washington University in Saint Louis, 660 South Euclid Ave, Box 8225, St. Louis, MO, 63110, USA
| | - Patrick L Hill
- Department of Psychological and Brain Sciences, Washington University in Saint Louis, One Booking Drive, St. Louis, MO, 63130, USA
| | - Thomas F Oltmanns
- Department of Psychological and Brain Sciences, Washington University in Saint Louis, One Booking Drive, St. Louis, MO, 63130, USA
| | - Janine D Bijsterbosch
- Department of Radiology, Washington University in Saint Louis, 660 South Euclid Ave, Box 8225, St. Louis, MO, 63110, USA
| | - Arpana Agrawal
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Alexander S Hatoum
- Department of Psychological and Brain Sciences, Washington University in Saint Louis, One Booking Drive, St. Louis, MO, 63130, USA
| | - Ryan Bogdan
- Department of Psychological and Brain Sciences, Washington University in Saint Louis, One Booking Drive, St. Louis, MO, 63130, USA.
| |
Collapse
|
21
|
Ferrari-Souza JP, Lussier FZ, Leffa DT, Therriault J, Tissot C, Bellaver B, Ferreira PC, Malpetti M, Wang YT, Povala G, Benedet AL, Ashton NJ, Chamoun M, Servaes S, Bezgin G, Kang MS, Stevenson J, Rahmouni N, Pallen V, Poltronetti NM, O’Brien JT, Rowe JB, Cohen AD, Lopez OL, Tudorascu DL, Karikari TK, Klunk WE, Villemagne VL, Soucy JP, Gauthier S, Souza DO, Zetterberg H, Blennow K, Zimmer ER, Rosa-Neto P, Pascoal TA. APOEε4 associates with microglial activation independently of Aβ plaques and tau tangles. SCIENCE ADVANCES 2023; 9:eade1474. [PMID: 37018391 PMCID: PMC10075966 DOI: 10.1126/sciadv.ade1474] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 03/02/2023] [Indexed: 06/01/2023]
Abstract
Animal studies suggest that the apolipoprotein E ε4 (APOEε4) allele is a culprit of early microglial activation in Alzheimer's disease (AD). Here, we tested the association between APOEε4 status and microglial activation in living individuals across the aging and AD spectrum. We studied 118 individuals with positron emission tomography for amyloid-β (Aβ; [18F]AZD4694), tau ([18F]MK6240), and microglial activation ([11C]PBR28). We found that APOEε4 carriers presented increased microglial activation relative to noncarriers in early Braak stage regions within the medial temporal cortex accounting for Aβ and tau deposition. Furthermore, microglial activation mediated the Aβ-independent effects of APOEε4 on tau accumulation, which was further associated with neurodegeneration and clinical impairment. The physiological distribution of APOE mRNA expression predicted the patterns of APOEε4-related microglial activation in our population, suggesting that APOE gene expression may regulate the local vulnerability to neuroinflammation. Our results support that the APOEε4 genotype exerts Aβ-independent effects on AD pathogenesis by activating microglia in brain regions associated with early tau deposition.
Collapse
Affiliation(s)
- João Pedro Ferrari-Souza
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Firoza Z. Lussier
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Douglas T. Leffa
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- ADHD Outpatient Program and Development Psychiatry Program, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Cécile Tissot
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Bruna Bellaver
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | - Maura Malpetti
- Department of Clinical Neurosciences, Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, UK
| | - Yi-Ting Wang
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Guilherme Povala
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Andréa L. Benedet
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Nicholas J. Ashton
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Mira Chamoun
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Stijn Servaes
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Gleb Bezgin
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Min Su Kang
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
- Artificial Intelligence and Computational Neurosciences lab, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
- LC Campbell Cognitive Neurology Unit, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Jenna Stevenson
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Nesrine Rahmouni
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Vanessa Pallen
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Nina Margherita Poltronetti
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - John T. O’Brien
- Department of Clinical Neurosciences, Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, UK
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - James B. Rowe
- Department of Clinical Neurosciences, Cambridge University Hospitals NHS Trust, University of Cambridge, Cambridge, UK
- MRC Cognition and Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Ann D. Cohen
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Oscar L. Lopez
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dana L. Tudorascu
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Thomas K. Karikari
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - William E. Klunk
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Jean-Paul Soucy
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Diogo O. Souza
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Eduardo R. Zimmer
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Graduate Program in Biological Sciences: Pharmacology and Therapeuctis, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer’s Disease Research Unit, Douglas Research Institute, Le Centre intégré universitaire de santé et de services sociaux (CIUSSS) de l'Ouest-de-l'Île-de-Montréal; Department of Neurology and Neurosurgery, Psychiatry and Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Tharick A. Pascoal
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
22
|
West GL, Patai ZE, Coutrot A, Hornberger M, Bohbot VD, Spiers HJ. Landmark-dependent Navigation Strategy Declines across the Human Life-Span: Evidence from Over 37,000 Participants. J Cogn Neurosci 2023; 35:452-467. [PMID: 36603038 DOI: 10.1162/jocn_a_01956] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Humans show a remarkable capacity to navigate various environments using different navigation strategies, and we know that strategy changes across the life span. However, this observation has been based on studies of small sample sizes. To this end, we used a mobile app-based video game (Sea Hero Quest) to test virtual navigation strategies and memory performance within a distinct radial arm maze level in over 37,000 participants. Players were presented with six pathways (three open and three closed) and were required to navigate to the three open pathways to collect a target. Next, all six pathways were made available and the player was required to visit the pathways that were previously unavailable. Both reference memory and working memory errors were calculated. Crucially, at the end of the level, the player was asked a multiple-choice question about how they found the targets (i.e., a counting-dependent strategy vs. a landmark-dependent strategy). As predicted from previous laboratory studies, we found the use of landmarks declined linearly with age. Those using landmark-based strategies also performed better on reference memory than those using a counting-based strategy. These results extend previous observations in the laboratory showing a decreased use of landmark-dependent strategies with age.
Collapse
Affiliation(s)
| | - Zita Eva Patai
- University College London, United Kingdom.,King's College London, United Kingdom
| | | | | | | | | |
Collapse
|
23
|
Li W, Li R, Yan S, Zhao Z, Shan Y, Qi Z, Lu J. Effect of APOE ε4 genotype on amyloid-β, glucose metabolism, and gray matter volume in cognitively normal individuals and amnestic mild cognitive impairment. Eur J Neurol 2023; 30:587-596. [PMID: 36448771 PMCID: PMC10107141 DOI: 10.1111/ene.15656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/21/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022]
Abstract
BACKGROUND AND PURPOSE The presence of apolipoprotein E ε4 (APOE ε4) is associated with an increased risk of developing Alzheimer disease (AD). The aim of this study was to assess the effects of APOE ε4 on amyloid-β (Aβ) pathology, glucose metabolism, and gray matter (GM) volume and their longitudinal changes in healthy control (HC) and amnestic mild cognitive impairment (aMCI). METHODS We included 50 HCs and 109 aMCI patients from the Alzheimer's Disease Neuroimaging Initiative phase 2/GO based on availability of baseline T1-weighted magnetic resonance imaging, 18 F-florbetapir positron emission tomography (PET), and 18 F-fluorodeoxyglucose (FDG) PET. Of these, 35 HCs and 67 aMCI patients who underwent 24-month scans were included for follow-up study. RESULTS Voxelwise analysis revealed that APOE ε4 carriers exhibited greater baseline Aβ deposition than APOE ε4 noncarriers in both diagnostic groups. However, there was no significant difference between APOE ε4 noncarriers and APOE ε4 carriers in terms of 18 F-FDG PET standardized uptake value ratio and GM volume. Region of interest-based analysis showed statistically significant greater Aβ deposition in APOE ε4 carriers than APOE ε4 noncarriers only in aMCI patients. Furthermore, APOE ε4 carriers generally exhibited a greater magnitude and spatial extent of longitudinal changes in Aβ deposition than APOE ε4 noncarriers in both diagnostic groups. CONCLUSIONS Our findings suggest a differential effect of APOE ε4 on Aβ pathology, glucose metabolism, and GM volume. Studying APOE ε4-related brain changes with neuroimaging biomarkers in preclinical AD offers an opportunity to further our understanding of the pathophysiology of AD at an early stage.
Collapse
Affiliation(s)
- Weihua Li
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | | | - Shaozhen Yan
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Zhilian Zhao
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Yi Shan
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Zhigang Qi
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Jie Lu
- Department of Radiology and Nuclear Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | | |
Collapse
|
24
|
Rogojin A, Gorbet DJ, Hawkins KM, Sergio LE. Differences in structural MRI and diffusion tensor imaging underlie visuomotor performance declines in older adults with an increased risk for Alzheimer's disease. Front Aging Neurosci 2023; 14:1054516. [PMID: 36711200 PMCID: PMC9877535 DOI: 10.3389/fnagi.2022.1054516] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/26/2022] [Indexed: 01/13/2023] Open
Abstract
Introduction Visuomotor impairments have been demonstrated in preclinical AD in individuals with a positive family history of dementia and APOE e4 carriers. Previous behavioral findings have also reported sex-differences in performance of visuomotor tasks involving a visual feedback reversal. The current study investigated the relationship between grey and white matter changes and non-standard visuomotor performance, as well as the effects of APOE status, family history of dementia, and sex on these brain-behavior relationships. Methods Older adults (n = 49) with no cognitive impairments completed non-standard visuomotor tasks involving a visual feedback reversal, plane-change, or combination of the two. Participants with a family history of dementia or who were APOE e4 carriers were considered at an increased risk for AD. T1-weighted anatomical scans were used to quantify grey matter volume and thickness, and diffusion tensor imaging measures were used to quantify white matter integrity. Results In APOE e4 carriers, grey and white matter structural measures were associated with visuomotor performance. Regression analyses showed that visuomotor deficits were predicted by lower grey matter thickness and volume in areas of the medial temporal lobe previously implicated in visuomotor control (entorhinal and parahippocampal cortices). This finding was replicated in the diffusion data, where regression analyses revealed that lower white matter integrity (lower FA, higher MD, higher RD, higher AxD) was a significant predictor of worse visuomotor performance in the forceps minor, forceps major, cingulum, inferior fronto-occipital fasciculus (IFOF), inferior longitudinal fasciculus (ILF), superior longitudinal fasciculus (SLF), and uncinate fasciculus (UF). Some of these tracts overlap with those important for visuomotor integration, namely the forceps minor, forceps major, SLF, IFOF, and ILF. Conclusion These findings suggest that measuring the dysfunction of brain networks underlying visuomotor control in early-stage AD may provide a novel behavioral target for dementia risk detection that is easily accessible, non-invasive, and cost-effective. The results also provide insight into the structural differences in inferior parietal lobule that may underlie previously reported sex-differences in performance of the visual feedback reversal task.
Collapse
Affiliation(s)
- Alica Rogojin
- School of Kinesiology and Health Science, York University, Toronto, ON, Canada,Centre for Vision Research, York University, Toronto, ON, Canada,Vision: Science to Applications (VISTA) Program, York University, Toronto, ON, Canada
| | - Diana J. Gorbet
- School of Kinesiology and Health Science, York University, Toronto, ON, Canada,Centre for Vision Research, York University, Toronto, ON, Canada
| | - Kara M. Hawkins
- School of Kinesiology and Health Science, York University, Toronto, ON, Canada
| | - Lauren E. Sergio
- School of Kinesiology and Health Science, York University, Toronto, ON, Canada,Centre for Vision Research, York University, Toronto, ON, Canada,*Correspondence: Lauren E. Sergio, ✉
| |
Collapse
|
25
|
Abstract
Memory impairment occurs in over a third of patients after symptomatic stroke. Memory deficits rarely occur in isolation but are an important component of the poststroke cognitive syndrome because of the strong relationship with the risk of poststroke dementia. In this review, we summarize available data on impairment of episodic memory, with a particular emphasis on the natural history of memory impairment after stroke and the factors influencing trajectory informed by an updated systematic review. We next discuss the pathophysiology of memory impairment and mechanisms of both decline and recovery of function. We then turn to the practical issue of measurement of memory deficits after stroke, emerging biomarkers, and therapeutic approaches. Our review identifies critical gaps, particularly in studies of the natural history that properly map the long-term trajectory of memory and the associations with factors that modulate prognosis. Few studies have used advanced neuroimaging and this, in conjunction with other biomarker approaches, has the potential to provide a much richer understanding of the mechanisms at play and promising therapeutic avenues.
Collapse
Affiliation(s)
- Michael J O'Sullivan
- Institute for Molecular Bioscience, University of Queensland, St Lucia, Australia (M.J.O.).,UQ Centre for Clinical Research, Faculty of Medicine, University of Queensland, Herston, Australia (M.J.O., X.L., D.G.).,Department of Neurology, Royal Brisbane and Women's Hospital, QLD, Australia (M.J.O.)
| | - Xuqian Li
- UQ Centre for Clinical Research, Faculty of Medicine, University of Queensland, Herston, Australia (M.J.O., X.L., D.G.)
| | - Dana Galligan
- UQ Centre for Clinical Research, Faculty of Medicine, University of Queensland, Herston, Australia (M.J.O., X.L., D.G.)
| | - Sarah T Pendlebury
- Wolfson Centre for Prevention of Stroke and Dementia, Nuffield Department of Clinical Neurosciences, University of Oxford, United Kingdom (S.T.P.).,Departments of Medicine and Geratology and UK National Institute for Health and Care Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, United Kingdom (S.T.P.)
| |
Collapse
|
26
|
Weerasekera A, Ion‐Mărgineanu A, Green C, Mody M, Nolan GP. Predictive models demonstrate age-dependent association of subcortical volumes and cognitive measures. Hum Brain Mapp 2022; 44:801-812. [PMID: 36222055 PMCID: PMC9842902 DOI: 10.1002/hbm.26100] [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: 06/29/2022] [Revised: 08/16/2022] [Accepted: 09/02/2022] [Indexed: 01/25/2023] Open
Abstract
Whether brain matter volume is correlated with cognitive functioning and higher intelligence is controversial. We explored this relationship by analysis of data collected on 193 healthy young and older adults through the "Leipzig Study for Mind-Body-Emotion Interactions" (LEMON) study. Our analysis involved four cognitive measures: fluid intelligence, crystallized intelligence, cognitive flexibility, and working memory. Brain subregion volumes were determined by magnetic resonance imaging. We normalized each subregion volume to the estimated total intracranial volume and conducted training simulations to compare the predictive power of normalized volumes of large regions of the brain (i.e., gray matter, cortical white matter, and cerebrospinal fluid), normalized subcortical volumes, and combined normalized volumes of large brain regions and normalized subcortical volumes. Statistical tests showed significant differences in the performance accuracy and feature importance of the subregion volumes in predicting cognitive skills for young and older adults. Random forest feature selection analysis showed that cortical white matter was the key feature in predicting fluid intelligence in both young and older adults. In young adults, crystallized intelligence was best predicted by caudate nucleus, thalamus, pallidum, and nucleus accumbens volumes, whereas putamen, amygdala, nucleus accumbens, and hippocampus volumes were selected for older adults. Cognitive flexibility was best predicted by the caudate, nucleus accumbens, and hippocampus in young adults and caudate and amygdala in older adults. Finally, working memory was best predicted by the putamen, pallidum, and nucleus accumbens in the younger group, whereas amygdala and hippocampus volumes were predictive in the older group. Thus, machine learning predictive models demonstrated an age-dependent association between subcortical volumes and cognitive measures. These approaches may be useful in predicting the likelihood of age-related cognitive decline and in testing of approaches for targeted improvement of cognitive functioning in older adults.
Collapse
Affiliation(s)
- Akila Weerasekera
- Department of Radiology, Athinoula A. Martinos Center for Biomedical ImagingMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | | | - Christopher Green
- Department of Diagnostic RadiologyDetroit Medical Center & Wayne State School of MedicineDetroitMichiganUSA
| | - Maria Mody
- Department of Radiology, Athinoula A. Martinos Center for Biomedical ImagingMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Garry P. Nolan
- Department of Microbiology & ImmunologyStanford University School of MedicinePalo AltoCaliforniaUSA
| |
Collapse
|
27
|
Zhang Z, Wu Y, Xiong D, Ibrahim JG, Srivastava A, Zhu H. LESA: Longitudinal Elastic Shape Analysis of Brain Subcortical Structures. J Am Stat Assoc 2022; 118:3-17. [PMID: 37153845 PMCID: PMC10162479 DOI: 10.1080/01621459.2022.2102984] [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: 10/04/2021] [Revised: 07/01/2022] [Accepted: 07/09/2022] [Indexed: 10/17/2022]
Abstract
Over the past 30 years, magnetic resonance imaging has become a ubiquitous tool for accurately visualizing the change and development of the brain's subcortical structures (e.g., hippocampus). Although subcortical structures act as information hubs of the nervous system, their quantification is still in its infancy due to many challenges in shape extraction, representation, and modeling. Here, we develop a simple and efficient framework of longitudinal elastic shape analysis (LESA) for subcortical structures. Integrating ideas from elastic shape analysis of static surfaces and statistical modeling of sparse longitudinal data, LESA provides a set of tools for systematically quantifying changes of longitudinal subcortical surface shapes from raw structure MRI data. The key novelties of LESA include: (i) it can efficiently represent complex subcortical structures using a small number of basis functions and (ii) it can accurately delineate the spatiotemporal shape changes of the human subcortical structures. We applied LESA to analyze three longitudinal neuroimaging data sets and showcase its wide applications in estimating continuous shape trajectories, building life-span growth patterns, and comparing shape differences among different groups. In particular, with the Alzheimer's Disease Neuroimaging Initiative (ADNI) data, we found that the Alzheimer's Disease (AD) can significantly speed the shape change of ventricle and hippocampus from 60 to 75 years old compared with normal aging.
Collapse
Affiliation(s)
- Zhengwu Zhang
- Department of Statistics and Operations Research, University of North Carolina at Chapel Hill Chapel Hill, North Carolina
| | - Yuexuan Wu
- Department of Statistics, Florida State University, Tallahassee, Florida
| | - Di Xiong
- Departments of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Joseph G. Ibrahim
- Departments of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Anuj Srivastava
- Department of Statistics, Florida State University, Tallahassee, Florida
| | - Hongtu Zhu
- Department of Statistics and Operations Research, University of North Carolina at Chapel Hill Chapel Hill, North Carolina
- Departments of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Departments of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Departments of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Biomedical Research Imaging Center, University of North Carolina at Chapel, Hill Chapel Hill, North Carolina
| |
Collapse
|
28
|
Shen B, Hernandez DG, Chitrala KN, Fanelli-Kuczmarski MT, Noren Hooten N, Pacheco NL, Mode NA, Zonderman AB, Ezike N, Evans MK. APOE gene region methylation is associated with cognitive performance in middle-aged urban adults. Neurobiol Aging 2022; 116:41-48. [PMID: 35561457 PMCID: PMC10878469 DOI: 10.1016/j.neurobiolaging.2022.03.010] [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: 05/28/2021] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 11/17/2022]
Abstract
Apolipoprotein (APOE) ε4 allele is a strong risk factor for Alzheimer's disease (AD) and cognitive decline. Epigenetic modifications such as DNA methylation (DNAm) play a central role in cognition. This study sought to identify DNAm sites in the APOE genomic region associated with cognitive performance in a racially diverse middle-aged cohort (n = 411). Cognitive performance was measured by 11 standard neuropsychological tests. Two CpG sites were associated with the Card Rotation and Benton Visual Retention cognitive tests. The methylation level of the CpG site cg00397545 was associated with Card Rotation Test score (p = 0.000177) and a novel CpG site cg10178308 was associated with Benton Visual Retention Test score (p = 0.000084). Significant associations were observed among the dietary inflammatory index, which reflects the inflammatory potential of the diet, cognitive performance and the methylation level of several CpG sites. Our results indicate that DNAm in the APOE genomic area is correlated with cognitive performance and may presage cognitive decline.
Collapse
Affiliation(s)
- Botong Shen
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Dena G Hernandez
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Kumaraswamy Naidu Chitrala
- Fels Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Marie T Fanelli-Kuczmarski
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Natasha L Pacheco
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Nicolle A Mode
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Ngozi Ezike
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
29
|
Badea A, Li D, Niculescu AR, Anderson RJ, Stout JA, Williams CL, Colton CA, Maeda N, Dunson DB. Absolute Winding Number Differentiates Mouse Spatial Navigation Strategies With Genetic Risk for Alzheimer's Disease. Front Neurosci 2022; 16:848654. [PMID: 35784847 PMCID: PMC9247395 DOI: 10.3389/fnins.2022.848654] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
Spatial navigation and orientation are emerging as promising markers for altered cognition in prodromal Alzheimer's disease, and even in cognitively normal individuals at risk for Alzheimer's disease. The different APOE gene alleles confer various degrees of risk. The APOE2 allele is considered protective, APOE3 is seen as control, while APOE4 carriage is the major known genetic risk for Alzheimer's disease. We have used mouse models carrying the three humanized APOE alleles and tested them in a spatial memory task in the Morris water maze. We introduce a new metric, the absolute winding number, to characterize the spatial search strategy, through the shape of the swim path. We show that this metric is robust to noise, and works for small group samples. Moreover, the absolute winding number better differentiated APOE3 carriers, through their straighter swim paths relative to both APOE2 and APOE4 genotypes. Finally, this novel metric supported increased vulnerability in APOE4 females. We hypothesized differences in spatial memory and navigation strategies are linked to differences in brain networks, and showed that different genotypes have different reliance on the hippocampal and caudate putamen circuits, pointing to a role for white matter connections. Moreover, differences were most pronounced in females. This departure from a hippocampal centric to a brain network approach may open avenues for identifying regions linked to increased risk for Alzheimer's disease, before overt disease manifestation. Further exploration of novel biomarkers based on spatial navigation strategies may enlarge the windows of opportunity for interventions. The proposed framework will be significant in dissecting vulnerable circuits associated with cognitive changes in prodromal Alzheimer's disease.
Collapse
Affiliation(s)
- Alexandra Badea
- Department of Radiology, Duke University, Durham, NC, United States
- Department of Neurology, Duke University, Durham, NC, United States
- Brain Imaging and Analysis Center, Duke University, Durham, NC, United States
- Biomedical Engineering, Duke University, Durham, NC, United States
| | - Didong Li
- Department of Computer Science, Princeton University, Princeton, NJ, United States
- Department of Biostatistics, University of California, Los Angeles, Los Angeles, CA, United States
| | | | | | - Jacques A. Stout
- Brain Imaging and Analysis Center, Duke University, Durham, NC, United States
| | - Christina L. Williams
- Department of Psychology and Neuroscience, Duke University, Durham, NC, United States
| | - Carol A. Colton
- Department of Neurology, Duke University, Durham, NC, United States
| | - Nobuyo Maeda
- Department of Pathology and Laboratory Medicine, The University of North Carolina, Chapel Hill, Chapel Hill, NC, United States
| | - David B. Dunson
- Department of Statistical Science, Duke University, Durham, NC, United States
| |
Collapse
|
30
|
Su Y, Zheng Q, Zhu L, Gu X, Lu J, Li L. Functions and underlying mechanisms of miR-650 in human cancers. Cancer Cell Int 2022; 22:132. [PMID: 35331235 PMCID: PMC8944108 DOI: 10.1186/s12935-022-02551-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 03/13/2022] [Indexed: 01/27/2023] Open
Abstract
MicroRNAs (miRNAs) are one type of noncoding RNAs that interfere with mRNA translation to downregulate gene expression, which results in posttranscriptional gene silencing. Over the past two decades, miRNAs have been widely reported to impact the progression of malignant tumours by interfering with cancer initiation and progression; therefore, miRNAs represent potential new diagnostic and therapeutic tools. miR-650 is a newly identified miR, and increasing studies have demonstrated that miR-650 plays critical roles in cancer progression, such as mediating the Wnt signalling pathway/AXIN1 (axis inhibition protein 1) axis in hepatocellular carcinoma. Nevertheless, associations between the expression patterns and molecular mechanisms of miR-650 in cancer have not been comprehensively described. In this article, we review the existing evidence regarding the mechanisms by which miR-650 expression is altered and their relation to cancer. Moreover, the promising clinical application of miR-650 for diagnosis and treatment is highlighted.
Collapse
Affiliation(s)
- Yuanshuai Su
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, National Clinical Research Center for Infectious Diseases, Zhejiang University, Hangzhou, 310003, China
| | - Qiuxian Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, National Clinical Research Center for Infectious Diseases, Zhejiang University, Hangzhou, 310003, China
| | - Lingxiao Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, National Clinical Research Center for Infectious Diseases, Zhejiang University, Hangzhou, 310003, China
| | - Xinyu Gu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, National Clinical Research Center for Infectious Diseases, Zhejiang University, Hangzhou, 310003, China
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, National Clinical Research Center for Infectious Diseases, Zhejiang University, Hangzhou, 310003, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, National Clinical Research Center for Infectious Diseases, Zhejiang University, Hangzhou, 310003, China.
| |
Collapse
|
31
|
Associations of circulating C-reactive proteins, APOE ε4, and brain markers for Alzheimer's disease in healthy samples across the lifespan. Brain Behav Immun 2022; 100:243-253. [PMID: 34920091 DOI: 10.1016/j.bbi.2021.12.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 11/12/2021] [Accepted: 12/11/2021] [Indexed: 12/14/2022] Open
Abstract
The apolipoprotein E gene ε4 allele (APOE ε4) and higher circulating level of C-reactive protein (CRP) have been extensively investigated as risk factors for Alzheimer's disease (AD). Paradoxically, APOE ε4 has been associated with lower levels of blood CRP in middle-aged and older populations. However, few studies have investigated this intriguing relation and its impact on neurological markers for AD in younger ages, nor across the whole lifespan. Here, we examine associations of blood CRP levels, APOE ε4, and biomarkers for AD in a cognitively healthy lifespan cohort (N up to 749; 20-81 years of age) and replicate the findings in UK Biobank (N = 304 322; 37-72 years of age), the developmental ABCD study (N = 10 283; 9-11 years of age), and a middle-aged sample (N = 339; 40-65 years of age). Hippocampal volume, brain amyloid-β (Aβ) plaque levels, cerebrospinal fluid (CSF) levels of Aβ and tau species, and neurofilament protein light protein (NFL) were used as AD biomarkers in subsamples. In addition, we examined the genetic contribution to the variation of CRP levels over different CRP ranges using polygenic scores for CRP (PGS-CRP). Our results show APOE ε4 consistently associates with low blood CRP levels across all age groups (p < 0.05). Strikingly, both ε4 and PGS-CRP associated mainly with blood CRP levels within the low range (<5mg/L). We then show both APOE ε4 and high CRP levels associate with smaller hippocampus volumes across the lifespan (p < 0.025). APOE ε4 was associated with high Aβ plaque levels in the brain (FDR-corrected p = 8.69x10-4), low levels of CSF Aβ42 (FDR-corrected p = 6.9x10-2), and lower ratios of Aβ42 to Aβ40 (FDR-corrected p = 5.08x10-5). Blood CRP levels were weakly correlated with higher ratio of CSF Aβ42 to Aβ40 (p = 0.03, FDR-corrected p = 0.4). APOE ε4 did not correlate with blood concentrations of another 9 inflammatory cytokines, and none of these cytokines correlated with AD biomarkers. CONCLUSION: The inverse correlation between APOEε 4 and blood CRP levels existed before any pathological AD biomarker was observed, and only in the low CRP level range. Thus, we suggest to investigate whether APOEε 4 can confer risk by being associated with a lower inflammatory response to daily exposures, possibly leading to greater accumulation of low-grade inflammatory stress throughout life. A lifespan perspective is needed to understand this relationship concerning risk of developing AD.
Collapse
|
32
|
Nir TM, Zhu AH, Gari IB, Dixon D, Islam T, Villalon-Reina JE, Medland SE, Thompson PM, Jahanshad N. Effects of ApoE4 and ApoE2 genotypes on subcortical magnetic susceptibility and microstructure in 27,535 participants from the UK Biobank. PACIFIC SYMPOSIUM ON BIOCOMPUTING. PACIFIC SYMPOSIUM ON BIOCOMPUTING 2022; 27:121-132. [PMID: 34890142 PMCID: PMC9009383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Disrupted iron homeostasis is associated with several neurodegenerative diseases, including Alzheimer's disease (AD), and may be partially modulated by genetic risk factors. Here we evaluated whether subcortical iron deposition is associated with ApoE genotype, which substantially affects risk for late-onset AD. We evaluated differences in subcortical quantitative susceptibility mapping (QSM), a type of MRI sensitive to cerebral iron deposition, between either ApoE4 (E3E4+E4E4) or ApoE2 (E2E3+E2E2) carriers and E3 homozygotes (E3E3) in 27,535 participants from the UK Biobank (age: 45-82 years). We found that ApoE4 carriers had higher hippocampal (d=0.036; p=0.012) and amygdalar (d=0.035; p=0.013) magnetic susceptibility, particularly individuals aged 65 years or older, while those carrying ApoE2 (which protects against AD) had higher QSM only in the hippocampus (d=0.05; p=0.006), particularly those under age 65. Secondary diffusion MRI microstructural associations in these regions revealed greater diffusivity and less diffusion restriction in E4 carriers, however no differences were detected in E2 carriers. Disease risk conferred by ApoE4 may be linked with higher subcortical iron burden in conjunction with inflammation or neuronal loss in aging individuals, while ApoE2 associations may not necessarily reflect unhealthy iron deposits earlier in life.
Collapse
Affiliation(s)
- Talia M Nir
- Imaging Genetics Center, Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina del Rey, California, 90292, USA,
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Suh H, Lee YM, Park JM, Lee BD, Moon E, Jeong H, Kim SY, Lee KY, Kim HJ, Pak K, Choi KU, Mun CW, Chung YI. Smaller hippocampal volume in APOE ε4 carriers independent of amyloid-β (Aβ) burden. Psychiatry Res Neuroimaging 2021; 317:111381. [PMID: 34508954 DOI: 10.1016/j.pscychresns.2021.111381] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 07/12/2021] [Accepted: 08/25/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE To investigate the association of the APOE ε4 genotype with hippocampal volume, independent of Aβ burden. METHOD This cross-sectional study included 71 participants with mild cognitive impairment or mild AD. All participants were divided into carriers or non-carriers of the ε4 allele. The main outcome was hippocampal volume measured using structural magnetic resonance imaging; 18F-florbetaben positron emission tomography was additionally performed to investigate the association of APOE ε4 genotype with hippocampal volumes, independently of Aβ burden. Analysis of covariance was conducted to compare the differences in hippocampal volumes between carriers and non-carriers of the ε4 allele after controlling for global Aβ burden or local hippocampal Aβ burden. RESULTS The APOE ε4 genotype was associated with a smaller right and total hippocampal volume (right: 3160.16 ± 365.71 vs. 3365.24 ± 434.88, p < 0.05; total: 6257.48 ± 790.60 vs. 6599.52 ± 840.58, p < 0.05), independent of Aβ burden. CONCLUSION Our findings on the association of APOEε4 genotype with hippocampal volume independent of Aβ burden suggest that the APOEε4 genotype may contribute to hippocampal neurodegeneration through an Aβ-independent mechanism.
Collapse
Affiliation(s)
- Hwagyu Suh
- Department of Psychiatry, Pusan National University School of Medicine, Busan, Republic of Korea; Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Young-Min Lee
- Department of Psychiatry, Pusan National University School of Medicine, Busan, Republic of Korea; Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea.
| | - Je-Min Park
- Department of Psychiatry, Pusan National University School of Medicine, Busan, Republic of Korea; Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Byung-Dae Lee
- Department of Psychiatry, Pusan National University School of Medicine, Busan, Republic of Korea; Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Eunsoo Moon
- Department of Psychiatry, Pusan National University School of Medicine, Busan, Republic of Korea; Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Hee Jeong
- Department of Psychiatry, Pusan National University School of Medicine, Busan, Republic of Korea; Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Soo Yeon Kim
- Department of Psychiatry, Pusan National University School of Medicine, Busan, Republic of Korea; Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Kang Yoon Lee
- Department of Psychiatry, Pusan National University School of Medicine, Busan, Republic of Korea; Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Hak-Jin Kim
- Department of Radiology, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Kyoungjune Pak
- Department of Nuclear Medicine, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Kyung-Un Choi
- Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea; Department of Pathology, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Chi-Woong Mun
- Department of Biomedical Engineering and FIRST, Inje University, Gimhae, Republic of Korea
| | - Young-In Chung
- Department of Psychiatry, Pusan National University School of Medicine, Yangsan, Republic of Korea
| |
Collapse
|
34
|
Forno G, Lladó A, Hornberger M. Going round in circles-The Papez circuit in Alzheimer's disease. Eur J Neurosci 2021; 54:7668-7687. [PMID: 34656073 DOI: 10.1111/ejn.15494] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 10/01/2021] [Accepted: 10/12/2021] [Indexed: 11/29/2022]
Abstract
The hippocampus is regarded as the pivotal structure for episodic memory symptoms associated with Alzheimer's disease (AD) pathophysiology. However, what is often overlooked is that the hippocampus is 'only' one part of a network of memory critical regions, the Papez circuit. Other Papez circuit regions are often regarded as less relevant for AD as they are thought to sit 'downstream' of the hippocampus. However, this notion is oversimplistic, and increasing evidence suggests that other Papez regions might be affected before or concurrently with the hippocampus. In addition, AD research has mostly focused on episodic memory deficits, whereas spatial navigation processes are also subserved by the Papez circuit with increasing evidence supporting its valuable potential as a diagnostic measure of incipient AD pathophysiology. In the current review, we take a step forward analysing recent evidence on the structural and functional integrity of the Papez circuit across AD disease stages. Specifically, we will review the integrity of specific Papez regions from at-genetic-risk (APOE4 carriers), to mild cognitive impairment (MCI), to dementia stage of sporadic AD and autosomal dominant AD (ADAD). We related those changes to episodic memory and spatial navigation/orientation deficits in AD. Finally, we provide an overview of how the Papez circuit is affected in AD diseases and their specific symptomology contributions. This overview strengthened the need for moving away from a hippocampal-centric view to a network approach on how the whole Papez circuit is affected in AD and contributes to its symptomology, informing future research and clinical approaches.
Collapse
Affiliation(s)
- Gonzalo Forno
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain.,School of Psychology, Universidad de los Andes, Santiago, Chile.,Neuropsychology and Clinical Neuroscience Laboratory (LANNEC), Physiopathology Department, ICBM, Neurosciences Department, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Albert Lladó
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | | |
Collapse
|
35
|
An N, Fu Y, Shi J, Guo HN, Yang ZW, Li YC, Li S, Wang Y, Yao ZJ, Hu B. Synergistic Effects of APOE and CLU May Increase the Risk of Alzheimer's Disease: Acceleration of Atrophy in the Volumes and Shapes of the Hippocampus and Amygdala. J Alzheimers Dis 2021; 80:1311-1327. [PMID: 33682707 DOI: 10.3233/jad-201162] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND The volume loss of the hippocampus and amygdala in non-demented individuals has been reported to increase the risk of developing Alzheimer's disease (AD). Many neuroimaging genetics studies mainly focused on the individual effects of APOE and CLU on neuroimaging to understand their neural mechanisms, whereas their synergistic effects have been rarely studied. OBJECTIVE To assess whether APOE and CLU have synergetic effects, we investigated the epistatic interaction and combined effects of the two genetic variants on morphological degeneration of hippocampus and amygdala in the non-demented elderly at baseline and 2-year follow-up. METHODS Besides the widely-used volume indicator, the surface-based morphometry method was also adopted in this study to evaluate shape alterations. RESULTS Our results showed a synergistic effect of homozygosity for the CLU risk allele C in rs11136000 and APOEɛ4 on the hippocampal and amygdalar volumes during a 2-year follow-up. Moreover, the combined effects of APOEɛ4 and CLU C were stronger than either of the individual effects in the atrophy progress of the amygdala. CONCLUSION These findings indicate that brain morphological changes are caused by more than one gene variant, which may help us to better understand the complex endogenous mechanism of AD.
Collapse
Affiliation(s)
- Na An
- School of Information Science and Engineering, Lanzhou University, Lanzhou, Gansu Province, China
| | - Yu Fu
- School of Information Science and Engineering, Lanzhou University, Lanzhou, Gansu Province, China
| | - Jie Shi
- School of Information Science and Engineering, Lanzhou University, Lanzhou, Gansu Province, China
| | - Han-Ning Guo
- School of Information Science and Engineering, Lanzhou University, Lanzhou, Gansu Province, China
| | - Zheng-Wu Yang
- School of Information Science and Engineering, Lanzhou University, Lanzhou, Gansu Province, China
| | - Yong-Chao Li
- School of Information Science and Engineering, Lanzhou University, Lanzhou, Gansu Province, China
| | - Shan Li
- School of Information Science and Engineering, Lanzhou University, Lanzhou, Gansu Province, China
| | - Yin Wang
- School of Information Science and Engineering, Lanzhou University, Lanzhou, Gansu Province, China
| | - Zhi-Jun Yao
- School of Information Science and Engineering, Lanzhou University, Lanzhou, Gansu Province, China
| | - Bin Hu
- School of Information Science and Engineering, Lanzhou University, Lanzhou, Gansu Province, China.,Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, Lanzhou, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | | |
Collapse
|
36
|
Apolipoprotein e (APOE) ε4 genotype influences memory performance following remote traumatic brain injury in U.S. military service members and veterans. Brain Cogn 2021; 154:105790. [PMID: 34487993 DOI: 10.1016/j.bandc.2021.105790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 08/27/2021] [Accepted: 08/29/2021] [Indexed: 10/20/2022]
Abstract
The purpose of this study was to examine the association between the apolipoprotein E (APOE) ε4 allele and neurocognitive functioning following traumatic brain injury (TBI) in military service members and veterans (SMVs). Participants included 176 SMVs with a history of remote TBI (≥1 year post-injury), categorized into mild (n = 100), moderate (n = 40), and severe (n = 36) TBI groups. Participants completed a neuropsychological assessment and APOE genotyping (n = 46 ε4+, n = 130 ε4-). Neurocognitive composite scores representing memory, executive functioning, and visual processing speed were computed. ANCOVAs adjusting for race, education, combat exposure, and PTSD symptom severity showed a significant main effect of ε4 on the memory composite, such that ε4+ SMVs exhibited poorer memory performance than ε4- SMVs. When ε2 allele carriers were removed from the analyses, associations with memory were strengthened, demonstrating a possible protective effect of the ε2 allele. No main effect of TBI group was identified on any cognitive composite, nor were there any significant TBI group × ε4 status interactions for any cognitive composite. Future studies with larger samples are needed to verify these findings, but our results suggest an important relationship between ε4 status and memory functioning following remote TBI of all severities.
Collapse
|
37
|
Schwarz C, Lange C, Benson GS, Horn N, Wurdack K, Lukas M, Buchert R, Wirth M, Flöel A. Severity of Subjective Cognitive Complaints and Worries in Older Adults Are Associated With Cerebral Amyloid-β Load. Front Aging Neurosci 2021; 13:675583. [PMID: 34408640 PMCID: PMC8365025 DOI: 10.3389/fnagi.2021.675583] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/22/2021] [Indexed: 01/19/2023] Open
Abstract
Subjective cognitive decline (SCD) is considered an early risk stage for dementia due to Alzheimer's disease (AD) and the development of pathological brain changes, such as the aggregation of amyloid-beta (amyloid-β) plaques. This study evaluates the association between specific features of SCD and cerebral amyloid-β load measured by positron emission tomography (PET) with 18F-florbetaben in 40 cognitively normal older individuals. Global amyloid-β, as well as regional amyloid-β load for the frontal, temporal, parietal, and cingulate cortex, was quantified. Specific features of SCD, such as subjective cognitive complaints and worry, were assessed using the 39-item Everyday Cognition Scales and the 16-item Penn State Worry Questionnaire. Spearman's rank partial correlation analyses, adjusted for age and apolipoprotein E ε4 status, were conducted to test the associations between specific features of SCD and cerebral amyloid-β load. The severity of subjective cognitive complaints in everyday memory and organization was positively correlated with amyloid-β load in the frontal cortex. In addition, the severity of subjective cognitive complaints in everyday planning was positively correlated with amyloid-β load in the parietal cortex. Higher levels of worry were associated with higher amyloid-β load in the frontal cortex. After correction of the PET data for partial volume effects, these associations were reduced to trend level. In conclusion, the severity of subjective cognitive complaints and the level of trait worry were positively associated with cortical amyloid-β burden, particularly in the frontal and parietal cortex. Further studies are required to elucidate the direction of these associations in order to develop strategies to prevent amyloid deposition and cognitive decline.
Collapse
Affiliation(s)
- Claudia Schwarz
- Department of Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Catharina Lange
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Gloria S Benson
- Department of Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nora Horn
- Department of Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Katharina Wurdack
- Department of Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Mathias Lukas
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Siemens Healthcare GmbH, Berlin, Germany
| | - Ralph Buchert
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Department for Diagnostic and Interventional Radiology and Nuclear Medicine, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Miranka Wirth
- German Center for Neurodegenerative Diseases (DZNE) Site: Dresden, Dresden, Germany
| | - Agnes Flöel
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany.,German Center for Neurodegenerative Diseases (DZNE) Site: Greifswald, Greifswald, Germany
| |
Collapse
|
38
|
Saeed U, Desmarais P, Masellis M. The APOE ε4 variant and hippocampal atrophy in Alzheimer's disease and Lewy body dementia: a systematic review of magnetic resonance imaging studies and therapeutic relevance. Expert Rev Neurother 2021; 21:851-870. [PMID: 34311631 DOI: 10.1080/14737175.2021.1956904] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Introduction: The apolipoprotein E ɛ4-allele (APOE-ɛ4) increases the risk not only for Alzheimer's disease (AD) but also for Parkinson's disease dementia and dementia with Lewy bodies (collectively, Lewy body dementia [LBD]). Hippocampal volume is an important neuroimaging biomarker for AD and LBD, although its association with APOE-ɛ4 is inconsistently reported. We investigated the association of APOE-ε4 with hippocampal atrophy quantified using magnetic resonance imaging in AD and LBD.Areas covered: Databases were searched for volumetric and voxel-based morphometric studies published up until December 31st, 2020. Thirty-nine studies (25 cross-sectional, 14 longitudinal) were included. We observed that (1) APOE-ε4 was associated with greater rate of hippocampal atrophy in longitudinal studies in AD and in those who progressed from mild cognitive impairment to AD, (2) association of APOE-ε4 with hippocampal atrophy in cross-sectional studies was inconsistent, (3) APOE-ɛ4 may influence hippocampal atrophy in dementia with Lewy bodies, although longitudinal investigations are needed. We comprehensively discussed methodological aspects, APOE-based therapeutic approaches, and the association of APOE-ε4 with hippocampal sub-regions and cognitive performance.Expert opinion: The role of APOE-ɛ4 in modulating hippocampal phenotypes may be further clarified through more homogenous, well-powered, and pathology-proven, longitudinal investigations. Understanding the underlying mechanisms will facilitate the development of prevention strategies targeting APOE-ɛ4.
Collapse
Affiliation(s)
- Usman Saeed
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Canada.,L.C. Campbell Cognitive Neurology Research Unit, Sunnybrook Health Sciences Centre, Toronto, Canada.,Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Canada
| | - Philippe Desmarais
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Canada.,L.C. Campbell Cognitive Neurology Research Unit, Sunnybrook Health Sciences Centre, Toronto, Canada.,Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Canada
| | - Mario Masellis
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Canada.,L.C. Campbell Cognitive Neurology Research Unit, Sunnybrook Health Sciences Centre, Toronto, Canada.,Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Canada.,Division of Neurology, Department of Medicine, University of Toronto, Toronto, Canada.,Cognitive and Movement Disorders Clinic, Sunnybrook Health Sciences Centre, Toronto, Canada
| |
Collapse
|
39
|
Piersson AD, Mohamad M, Suppiah S, Rajab NF. Topographical patterns of whole-brain structural alterations in association with genetic risk, cerebrospinal fluid, positron emission tomography biomarkers of Alzheimer’s disease, and neuropsychological measures. Clin Transl Imaging 2021. [DOI: 10.1007/s40336-021-00440-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
40
|
Mentink LJ, Guimarães JPOFT, Faber M, Sprooten E, Olde Rikkert MGM, Haak KV, Beckmann CF. Functional co-activation of the default mode network in APOE ε4-carriers: A replication study. Neuroimage 2021; 240:118304. [PMID: 34329959 DOI: 10.1016/j.neuroimage.2021.118304] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 05/27/2021] [Accepted: 06/22/2021] [Indexed: 11/19/2022] Open
Abstract
Structural and functional alterations of the brain in persons genetically at-risk for Alzheimer's disease (AD) are crucial in unravelling AD development. Filippini et al. found that the default mode network (DMN) is already affected in young APOE ε4-carriers, with increased co-activation of the DMN during rest and increased hippocampal task activation. We aimed to replicate the early findings of Filippini et al, using the APOE gene, still the principal AD risk gene, and extended this with a polygenic risk score (PRS) analysis for AD, using the Human Connectome Project dataset (HCP). We included participants from the HCP S1200 dataset (age range: 22-36 years). We studied morphometric features, functional DMN co-activation and functional task activation of recollection performance. Permutation Analysis of Linear Models (PALM) was used to test for group differences between APOE ε4-carriers and non-carriers, and to test the association with PRS. PALM controls for biases induced by the family structure of the HCP sample. Results were family-wise error rate corrected at p < 0.05. Our primary analysis did not replicate the early findings of Filippini et al. (2009). However, compared with non-carriers, APOE ε4-carriers showed increased functional activation during the encoding of subsequently recollected items in areas related to facial recognition (p<0.05, t>756.11). This increased functional activation was also positively associated with PRS (APOE variants included) (p<0.05, t>647.55). Our results are supportive for none to limited genetic effects on brain structure and function in young adults. Taking the methodological considerations of replication studies into account, the true effect of APOE ε4-carriership is likely smaller than indicated in the Filippini paper. However, it still holds that we may not yet be able to detect already present measurable effects decades before a clinical expression of AD. Since the mechanistic pathway of AD is likely to encompass many different factors, further research should be focused on the interactions of genetic risk, biomarkers, aging and lifestyle factors over the life course. Sensitive functional neuroimaging as used here may help disentangling these complex interactions.
Collapse
Affiliation(s)
- Lara J Mentink
- Department of Geriatrics, Radboudumc Alzheimer Centre, Radboud University Medical Center, Nijmegen, The Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - João P O F T Guimarães
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Myrthe Faber
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Communication and Cognition, Tilburg Center for Cognition and Communication, Tilburg University, Tilburg, The Netherlands.
| | - Emma Sprooten
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Marcel G M Olde Rikkert
- Department of Geriatrics, Radboudumc Alzheimer Centre, Radboud University Medical Center, Nijmegen, The Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Koen V Haak
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Christian F Beckmann
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands; Centre for Functional MRI of the Brain (FMRIB), Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
41
|
Pardilla-Delgado E, Torrico-Teave H, Sanchez JS, Ramirez-Gomez LA, Baena A, Bocanegra Y, Vila-Castelar C, Fox-Fuller JT, Guzmán-Vélez E, Martínez J, Alvarez S, Ochoa-Escudero M, Lopera F, Quiroz YT. Associations between subregional thalamic volume and brain pathology in autosomal dominant Alzheimer's disease. Brain Commun 2021; 3:fcab101. [PMID: 34095834 PMCID: PMC8172494 DOI: 10.1093/braincomms/fcab101] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 02/01/2021] [Accepted: 03/19/2021] [Indexed: 12/02/2022] Open
Abstract
Histopathological reports suggest that subregions of the thalamus, which regulates multiple physiological and cognitive processes, are not uniformly affected by Alzheimer's disease. Despite this, structural neuroimaging studies often consider the thalamus as a single region. Identification of in vivo Alzheimer's-dependent volumetric changes in thalamic subregions may aid the characterization of early nuclei-specific neurodegeneration in Alzheimer's disease. Here, we leveraged access to the largest single-mutation cohort of autosomal-dominant Alzheimer's disease to test whether cross-sectional abnormalities in subregional thalamic volumes are evident in non-demented mutation carriers (n = 31), compared to non-carriers (n = 36), and whether subregional thalamic volume is associated with age, markers of brain pathology and cognitive performance. Using automatic parcellation we examined the thalamus in six subregions (anterior, lateral, ventral, intralaminar, medial, and posterior) and their relation to age and brain pathology (amyloid and tau), as measured by PET imaging. No between-group differences were observed in the volume of the thalamic subregions. In carriers, lower volume in the medial subregion was related to increased cortical amyloid and entorhinal tau burden. These findings suggest that thalamic Alzheimer's-related volumetric reductions are not uniform even in preclinical and prodromal stages of autosomal-dominant Alzheimer's disease and therefore, this structure should not be considered as a single, unitary structure in Alzheimer's disease research.
Collapse
Affiliation(s)
| | | | - Justin S Sanchez
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | | | - Ana Baena
- Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellin 050010, Colombia
| | - Yamile Bocanegra
- Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellin 050010, Colombia
| | - Clara Vila-Castelar
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Joshua T Fox-Fuller
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Boston University, Boston, MA 02215, USA
| | | | - Jairo Martínez
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | | | | | - Francisco Lopera
- Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellin 050010, Colombia
| | - Yakeel T Quiroz
- Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Grupo de Neurociencias de Antioquia, Universidad de Antioquia, Medellin 050010, Colombia
| |
Collapse
|
42
|
Bussy A, Patel R, Plitman E, Tullo S, Salaciak A, Bedford SA, Farzin S, Béland ML, Valiquette V, Kazazian C, Tardif CL, Devenyi GA, Chakravarty MM. Hippocampal shape across the healthy lifespan and its relationship with cognition. Neurobiol Aging 2021; 106:153-168. [PMID: 34280848 DOI: 10.1016/j.neurobiolaging.2021.03.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 03/02/2021] [Accepted: 03/29/2021] [Indexed: 01/18/2023]
Abstract
The study of the hippocampus across the healthy adult lifespan has rendered inconsistent findings. While volumetric measurements have often been a popular technique for analysis, more advanced morphometric techniques have demonstrated compelling results that highlight the importance and improved specificity of shape-based measures. Here, the MAGeT Brain algorithm was applied on 134 healthy individuals aged 18-81 years old to extract hippocampal subfield volumes and hippocampal shape measurements, namely: local surface area (SA) and displacement. We used linear-, second- or third-order natural splines to examine the relationships between hippocampal measures and age. In addition, partial least squares analyses were performed to relate volume and shape measurements with cognitive and demographic information. Volumetric results indicated a relative preservation of the right cornus ammonis 1 with age and a global volume reduction linked with older age, female sex, lower levels of education and cognitive performance. Vertex-wise analysis demonstrated an SA preservation in the anterior hippocampus with a peak during the sixth decade, while the posterior hippocampal SA gradually decreased across lifespan. Overall, SA decrease was linked to older age, female sex and, to a lesser extent lower levels of education and cognitive performance. Outward displacement in the lateral hippocampus and inward displacement in the medial hippocampus were enlarged with older age, lower levels of cognition and education, indicating an accentuation of the hippocampal "C" shape with age. Taken together, our findings suggest that vertex-wise analyses have higher spatial specifity and that sex, education, and cognition are implicated in the differential impact of age on hippocampal subregions throughout its anteroposterior and medial-lateral axes. This article is part of the Virtual Special Issue titled COGNITIVE NEU- ROSCIENCE OF HEALTHY AND PATHOLOGICAL AGING. The full issue can be found on ScienceDirect at https://www.sciencedirect.com/journal/neurobiology-of-aging/special-issue/105379XPWJP.
Collapse
Affiliation(s)
- Aurélie Bussy
- Computional Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada.
| | - Raihaan Patel
- Computional Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada
| | - Eric Plitman
- Computional Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Stephanie Tullo
- Computional Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Alyssa Salaciak
- Computional Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Saashi A Bedford
- Computional Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Sarah Farzin
- Computional Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Marie-Lise Béland
- Computional Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Vanessa Valiquette
- Computional Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Christina Kazazian
- Computional Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Christine L Tardif
- Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada; McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Gabriel A Devenyi
- Computional Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - M Mallar Chakravarty
- Computional Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada; Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
43
|
Chen Y, Li TR, Hao SW, Wang XN, Cai YN, Han Y. Blood NCAPH2 Methylation Is Associated With Hippocampal Volume in Subjective Cognitive Decline With Apolipoprotein E ε4 Non-carriers. Front Aging Neurosci 2021; 13:632382. [PMID: 33603659 PMCID: PMC7884760 DOI: 10.3389/fnagi.2021.632382] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 01/06/2021] [Indexed: 11/13/2022] Open
Abstract
Objective: This study assessed the methylation of peripheral NCAPH2 in individuals with subjective cognitive decline (SCD), identified its correlation with the hippocampal volume, and explored whether the correlation is influenced by apolipoprotein E ε4 (APOE ε4) status. Methods: Cognitively normal controls (NCs, n = 56), individuals with SCD (n = 81), and patients with objective cognitive impairment (OCI, n = 51) were included from the Sino Longitudinal Study on Cognitive Decline (NCT03370744). All participants completed neuropsychological assessments, blood tests, and structural MRI. NCAPH2 methylation was compared according to the diagnostic and APOE ε4 status. Partial correlation analysis was conducted to assess the correlations between the hippocampal volume, cognitive tests, and the NCAPH2 methylation levels. Results: Individuals with SCD and patients with OCI showed significantly lower levels of NCAPH2 methylation than NCs, which were independent of the APOE ε4 status. The NCAPH2 methylation levels and the hippocampal volumes were positively correlated in the SCD APOE ε4 non-carriers but not in the OCI group. No association was found between the NCAPH2 methylation levels and the cognitive function. Conclusion: Abnormal changes in blood NCAPH2 methylation were found to occur in SCD, indicating its potential to be used as a useful peripheral biomarker in the early stage of Alzheimer's disease screening.
Collapse
Affiliation(s)
- Ying Chen
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Department of Neurology, Zhejiang Taizhou Municipal Hospital, Taizhou, Zhejiang, China
| | - Tao-Ran Li
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Shu-Wen Hao
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Xiao-Ni Wang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Yan-Ning Cai
- Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Ying Han
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, China.,National Clinical Research Center for Geriatric Disorders, Beijing, China
| |
Collapse
|
44
|
Yee A, Tsui NBY, Kwan RYC, Leung AYM, Lai CKY, Chung T, Lau JYN, Fok M, Dai DLK, Lau LT. Apolipoprotein E Gene Revisited: Contribution of Rare Variants to Alzheimer's Disease Susceptibility in Southern Chinese. Curr Alzheimer Res 2021; 18:67-79. [PMID: 33761857 DOI: 10.2174/1567205018666210324111401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 12/28/2020] [Accepted: 02/16/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND APOE ε4 is the best-known risk factor for late-onset alzheimer's disease (AD). Population studies have demonstrated a relatively low prevalence of APOE ε4 among Chinese population, implying additional risk factors that are Chinese-specific may exist. Apart from - alleles, genetic variation profile along the full-length APOE has rarely been investigated. OBJECTIVE In this study, we filled this gap by comprehensively determining all genetic variations in APOE and investigated their potential associations with late-onset AD and mild cognitive impairment (MCI) in southern Chinese. METHODS Two hundred and fifty-seven southern Chinese participants were recruited, of whom 69 were AD patients, 83 had MCI, and 105 were normal controls. Full-length APOE from promoter to 3'UTR regions were sequenced. Genetic variants were identified and compared among the three groups. RESULTS While APOE ε4 was more significantly found in AD patients, the prevalence of APOE ε4 in southern Chinese AD patients was the lowest when compared to other areas of China and nearby regions, as well as other countries worldwide. We further identified 13 rare non-singleton variants in APOE. Significantly more AD patients carried any of the rare non-singleton variants than MCI and normal subjects. Such difference was observed in the non-carriers of ε4-allele only. Among the identified rare variants, the potential functional impact was predicted for rs532314089, rs553874843, rs533904656 and rs370594287. CONCLUSION Our study suggests an ethnic difference in genetic risk composition of AD in southern Chinese. Rare variants on APOE are a potential candidate for AD risk stratification biomarker in addition to APOE-ε4.
Collapse
Affiliation(s)
- Anita Yee
- Avalon Genomics (Hong Kong) Limited, Shatin,Hong Kong
| | | | - Rick Y C Kwan
- Centre for Gerontological Nursing, School of Nursing, The Hong Kong Polytechnic University, Hung Hom,Hong Kong
| | - Angela Y M Leung
- Centre for Gerontological Nursing, School of Nursing, The Hong Kong Polytechnic University, Hung Hom,Hong Kong
| | - Claudia K Y Lai
- Centre for Gerontological Nursing, School of Nursing, The Hong Kong Polytechnic University, Hung Hom,Hong Kong
| | - Teresa Chung
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom,Hong Kong
| | | | - Manson Fok
- Faculty of Health Sciences, Macau University of Science and Technology, Taipa, Macau,Hong Kong
| | - David L K Dai
- Hong Kong Alzheimer's Disease Association, Wang Tau Hom,Hong Kong
| | - Lok-Ting Lau
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom,Hong Kong
| |
Collapse
|
45
|
Martí‐Juan G, Sanroma‐Guell G, Cacciaglia R, Falcon C, Operto G, Molinuevo JL, González Ballester MÁ, Gispert JD, Piella G. Nonlinear interaction between APOE ε4 allele load and age in the hippocampal surface of cognitively intact individuals. Hum Brain Mapp 2021; 42:47-64. [PMID: 33017488 PMCID: PMC7721244 DOI: 10.1002/hbm.25202] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/16/2020] [Accepted: 08/11/2020] [Indexed: 01/27/2023] Open
Abstract
The ε4 allele of the gene Apolipoprotein E is the major genetic risk factor for Alzheimer's Disease. APOE ε4 has been associated with changes in brain structure in cognitively impaired and unimpaired subjects, including atrophy of the hippocampus, which is one of the brain structures that is early affected by AD. In this work we analyzed the impact of APOE ε4 gene dose and its association with age, on hippocampal shape assessed with multivariate surface analysis, in a ε4-enriched cohort of n = 479 cognitively healthy individuals. Furthermore, we sought to replicate our findings on an independent dataset of n = 969 individuals covering the entire AD spectrum. We segmented the hippocampus of the subjects with a multi-atlas-based approach, obtaining high-dimensional meshes that can be analyzed in a multivariate way. We analyzed the effects of different factors including APOE, sex, and age (in both cohorts) as well as clinical diagnosis on the local 3D hippocampal surface changes. We found specific regions on the hippocampal surface where the effect is modulated by significant APOE ε4 linear and quadratic interactions with age. We compared between APOE and diagnosis effects from both cohorts, finding similarities between APOE ε4 and AD effects on specific regions, and suggesting that age may modulate the effect of APOE ε4 and AD in a similar way.
Collapse
Affiliation(s)
- Gerard Martí‐Juan
- BCN MedTech, Departament de Tecnologies de la Informació i les ComunicacionsUniversitat Pompeu FabraBarcelonaSpain
| | | | - Raffaele Cacciaglia
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Medical Research Institute (IMIM)BarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
| | - Carles Falcon
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Medical Research Institute (IMIM)BarcelonaSpain
- Centro de Investigación Biomédica en Red de BioingenieríaBiomateriales y Nanomedicina (CIBERBBN)MadridSpain
| | - Grégory Operto
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Medical Research Institute (IMIM)BarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Medical Research Institute (IMIM)BarcelonaSpain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES)MadridSpain
- Universitat Pompeu FabraBarcelonaSpain
| | - Miguel Ángel González Ballester
- BCN MedTech, Departament de Tecnologies de la Informació i les ComunicacionsUniversitat Pompeu FabraBarcelonaSpain
- ICREABarcelonaSpain
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC)Pasqual Maragall FoundationBarcelonaSpain
- Hospital del Mar Medical Research Institute (IMIM)BarcelonaSpain
- Centro de Investigación Biomédica en Red de BioingenieríaBiomateriales y Nanomedicina (CIBERBBN)MadridSpain
- Universitat Pompeu FabraBarcelonaSpain
| | - Gemma Piella
- BCN MedTech, Departament de Tecnologies de la Informació i les ComunicacionsUniversitat Pompeu FabraBarcelonaSpain
| | | | | |
Collapse
|
46
|
Piers RJ, Liu Y, Ang TF, Tao Q, Au R, Qiu WQ. Association Between Elevated Depressive Symptoms and Cognitive Function Moderated by APOE4 Status: Framingham Offspring Study. J Alzheimers Dis 2021; 80:1269-1279. [PMID: 33646152 PMCID: PMC8172078 DOI: 10.3233/jad-200998] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Depression and Apolipoprotein E4 (APOE4) are associated with decreased cognitive function and differences in brain structure. OBJECTIVE This study investigated whether APOE4 status moderates the association between elevated depressive symptoms, cognitive function, and brain structure. METHODS Stroke- and dementia-free participants (n = 1,968) underwent neuropsychological evaluation, brain MRI, and depression screening. Linear and logistic regression was used to examine all associations. Secondary analyses were performed using interaction terms to assess effect modification by APOE4 status. RESULTS Elevated depressive symptoms were associated with lower cognitive performance in several domains. In stratified analyses, elevated depressive symptoms were associated with poorer visual short- and long-term memory performance for APOE4 + participants. Elevated depressive symptoms were not associated with any brain structure in this study sample. CONCLUSION Elevated depressive symptoms impact cognitive function in non-demented individuals. Having the APOE4 allele may exacerbate the deleterious effects of elevated depressive symptoms on visual memory performance. Screening for elevated depressive symptoms in both research studies and clinical practice may be warranted to avoid false positive identification of neurodegeneration, particularly among those who are APOE4 + .
Collapse
Affiliation(s)
- Ryan J. Piers
- Department of Psychological and Brain Sciences, Boston University, Boston, MA, USA
| | - Yulin Liu
- The Framingham Heart Study, Boston University School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Ting F.A. Ang
- The Framingham Heart Study, Boston University School of Medicine, Boston, MA, USA
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | - Qiushan Tao
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Rhoda Au
- The Framingham Heart Study, Boston University School of Medicine, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- Alzheimer’s Disease Center, Boston University School of Medicine, Boston, MA, USA
| | - Wendy Q. Qiu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
- Alzheimer’s Disease Center, Boston University School of Medicine, Boston, MA, USA
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
47
|
Harrison AT, McAllister T, McCrea M, Broglio SP, Moore RD. Recovery Profiles after Concussion among Male Student-Athletes and Service Cadets with a Family History of Neurodegenerative Disease: Data from the NCAA-DoD CARE Consortium. J Neurotrauma 2020; 38:485-492. [PMID: 33280495 DOI: 10.1089/neu.2020.7386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Preliminary evidence indicates that genetic factors associated with having a family history of neurodegenerative disease (fhNDD) may predispose an individual to persistent symptoms and poorer cognitive performance after concussion. No previous study, however, longitudinally examined athletes with (+) and without (-) a fhNDD. Therefore, we aimed to compare clinical symptoms and cognitive performance of fhNDD+ and fhNDD- athletes at baseline and at multiple time points after concussion. Questionnaire data from the Concussion Assessment, Research and Education (CARE) Consortium were used to identify male athletes and cadets with (n = 51) and without (n = 102) a fhNDD (Alzheimer disease, Parkinson disease, mild cognitive impairment, and non-Alzheimer dementia). All athletes completed the SCAT3 symptom checklist and ImPACT test before their sport season and again within 24-48 h of injury, at the unrestricted return-to-play, and at six months post-concussion. Compared with fhNDD-, fhNDD+ individuals demonstrated greater decrements in visual memory (relative to baseline) 24-48 h post-injury (p < 0.05, d = 0.18). In addition, a main effect of group was observed for impulse control. Compared with fhNDD- athletes, fhNDD+ individuals demonstrated greater decrements in impulse control, 24-48 h post-injury, at the return to play, and at six-month assessments (p < 0.01, d = 0.23). These findings suggest that male athletes with a fhNDD may exhibit greater decrements in cognitive performance after concussion. Small, subtle deficits in cognitive performance may still significantly hinder day-to-day function in student-athletes.
Collapse
Affiliation(s)
- Adam T Harrison
- Department of Exercise Science, University of South Carolina, Columbia, South Carolina, USA
| | - Thomas McAllister
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Michael McCrea
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Steven P Broglio
- Michigan Concussion Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Robert D Moore
- Department of Exercise Science, University of South Carolina, Columbia, South Carolina, USA
| | | |
Collapse
|
48
|
Soares JZ, Pettersen R, Benth JŠ, Persson K, Strobel C, Selbæk G, Bogdanovic N. Vitamin D Levels, APOE Allele, and MRI Volumetry Assessed by NeuroQuant in Norwegian Adults with Cognitive Symptoms. J Alzheimers Dis 2020; 79:311-321. [PMID: 33252081 DOI: 10.3233/jad-201018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Allele ɛ4 of the apolipoprotein (APOE∈4) gene is the strongest known genetic risk factor for late-onset sporadic Alzheimer's disease. A possible relationship between vitamin D and APOE is not yet clear. OBJECTIVE In this exploratory, cross-sectional study, we examined the association between serum levels of 25-hydroxyvitamin D [25(OH)D] and brain volumes and the associations of both serum levels of 25(OH)D and APOE polymorphism to brain volumes in 127 persons (mean age 66 years) with cognitive symptoms. METHODS All subjects were examined with fully automated software for MRI volumetry, NeuroQuant. RESULTS After adjustment for relevant covariates, higher serum 25(OH)D levels were associated with greater volumes of cortical gray matter on both left (p = 0.02) and right (p = 0.04) sides. When both 25(OH)D levels and APOE genotype were used as the main covariates, no significant associations were found between vitamin D level and brain volume in any of the 11 brain regions. In adjusted models, only homozygous but not heterozygous APOE∈4 allele carriers had significantly larger inferior lateral ventricles (p = 0.003) and smaller hippocampal volume (p = 0.035) than those without ɛ4. Homozygous APOE∈4 carriers also had significantly higher vitamin D levels (p = 0.009) compared to persons without the APOE∈4 allele. CONCLUSION Higher vitamin D levels might have a preserving effect on cortical grey matter volume.
Collapse
Affiliation(s)
- Jelena Zugic Soares
- Medical Department, Section of Geriatrics, Lovisenberg Diaconal Hospital, Nydalen, Oslo, Norway
| | - Renate Pettersen
- Medical Department, Section of Geriatrics, Lovisenberg Diaconal Hospital, Nydalen, Oslo, Norway
| | - Jūratė Šaltytė Benth
- Institute of Clinical Medicine, Campus Ahus, University of Oslo, Blindern, Norway.,Health Services Research Unit, Akershus University Hospital, Lørenskog, Norway
| | - Karin Persson
- Department of Geriatric medicine, Oslo University Hospital, Nydalen, Oslo, Norway.,Norwegian National Advisory Unit on Ageing and Health, Vestfold Hospital Trust, Tønsberg, Norway
| | - Carsten Strobel
- Medical Department, Section of Geriatrics, Lovisenberg Diaconal Hospital, Nydalen, Oslo, Norway
| | - Geir Selbæk
- Department of Geriatric medicine, Oslo University Hospital, Nydalen, Oslo, Norway.,Norwegian National Advisory Unit on Ageing and Health, Vestfold Hospital Trust, Tønsberg, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Nenad Bogdanovic
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Karolinska Institutet, Department for Neurobiology, Caring Science and Society, Division of Clinical Geriatrics Novum Research Park, Stockholm, Sweden
| |
Collapse
|
49
|
Werden E, Khlif MS, Bird LJ, Cumming T, Bradshaw J, Khan W, Pase M, Restrepo C, Veldsman M, Egorova N, Patel SK, Gottlieb E, Brodtmann A. APOE ɛ4 Carriers Show Delayed Recovery of Verbal Memory and Smaller Entorhinal Volume in the First Year After Ischemic Stroke. J Alzheimers Dis 2020; 71:245-259. [PMID: 31381519 DOI: 10.3233/jad-190566] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND The apolipoprotein E (APOE) gene ɛ4 allele is a risk factor for Alzheimer's disease and cardiovascular disease. However, its relationship with cognition and brain volume after stroke is not clear. OBJECTIVE We compared cognition and medial temporal lobe volumes in APOEɛ4 carriers and non-carriers in the first year after ischemic stroke. METHODS We sampled 20 APOEɛ4 carriers and 20 non-carriers from a larger cohort of 135 ischemic stroke participants in the longitudinal CANVAS study. Participants were matched on a range of demographic and stroke characteristics. We used linear mixed-effect models to compare cognitive domain z-scores (attention, processing speed, executive function, verbal and visual memory, language, visuospatial function) and regional medial temporal lobe volumes (hippocampal, entorhinal cortex) between groups at each time-point (3, 12-months post-stroke), and within groups across time-points. APOE gene single nucleotide polymorphisms (SNPs; rs7412, rs429358) were genotyped on venous blood. RESULTS APOEɛ4 carriers and non-carriers did not differ on any demographic, clinical, or stroke variable. Carriers performed worse than non-carriers in verbal memory at 3 months post-stroke (p = 0.046), but were better in executive function at 12 months (p = 0.035). Carriers demonstrated a significant improvement in verbal memory (p = 0.012) and executive function (p = 0.015) between time-points. Non-carriers demonstrated a significant improvement in visual memory (p = 0.0005). Carriers had smaller bilateral entorhinal cortex volumes (p < 0.05), and larger right sided and contralesional hippocampal volumes, at both time-points (p < 0.05). CONCLUSION APOE ɛ4 is associated with delayed recovery of verbal memory function and reduced entorhinal cortex volumes in the first year after ischemic stroke.
Collapse
Affiliation(s)
- Emilio Werden
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Mohamed Salah Khlif
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Laura J Bird
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Toby Cumming
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | | | - Wasim Khan
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Matthew Pase
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Carolina Restrepo
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Michele Veldsman
- Department of Experimental Psychology, University of Oxford, Oxford, UK
| | - Natalia Egorova
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia.,Melbourne School of Psychological Sciences, University of Melbourne, Melbourne, Australia
| | - Sheila K Patel
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Elie Gottlieb
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Amy Brodtmann
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia.,Austin Health, Heidelberg, Melbourne, VIC, Australia.,Eastern Clinical Research Unit, Box Hill Hospital, Melbourne, VIC, Australia
| |
Collapse
|
50
|
Chao LL. The Prevalence of Mild Cognitive Impairment in a Convenience Sample of 202 Gulf War Veterans. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E7158. [PMID: 33007845 PMCID: PMC7579246 DOI: 10.3390/ijerph17197158] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/01/2020] [Accepted: 09/10/2020] [Indexed: 11/16/2022]
Abstract
Gulf War Illness (GWI) is a chronic, multisymptom disorder estimated to affect approximately 25-32% of Gulf War veterans (GWVs). Cognitive dysfunction is a common symptom of GWI. On the continuum of cognitive decline, mild cognitive impairment (MCI) is conceptualized as a transitional phase between normal aging and dementia. Individuals with MCI exhibit cognitive decline but have relatively spared activities of daily function and do not meet criteria for dementia. The current study sought to investigate the prevalence of MCI in a convenience sample of 202 GWVs (median age: 52 years; 18% female). Twelve percent of the sample (median age: 48 years) had MCI according to an actuarial neuropsychological criterion, a rate materially higher than expected for this age group. GWVs with MCI also had a smaller hippocampal volume and a thinner parietal cortex, higher rates of current posttraumatic stress disorder and major depressive disorder compared to GWVs without MCI. Because people with MCI are more likely to progress to dementia compared to those with normal cognition, these results may portend future higher rates of dementia among deployed GWVs.
Collapse
Affiliation(s)
- Linda L. Chao
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA 94143, USA;
- Department of Psychiatry and Behavioral Science, University of California, San Francisco, CA 94143, USA
- San Francisco Veterans Affairs Health Care System, San Francisco, CA 94121, USA
| |
Collapse
|