1
|
Gong C, Song W, Zhu Z, Yang D, Zhao X, Xu Y, Zhao H. APOE ε4 influences the dynamic functional connectivity variability and cognitive performance in Alzheimer's disease. J Alzheimers Dis 2025; 104:1103-1114. [PMID: 40151915 DOI: 10.1177/13872877251322687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
BackgroundApolipoprotein E (APOE) ε4 is the most significant genetic risk factor for sporadic Alzheimer's disease (AD). However, its impact on the dynamic changes in resting-state functional connectivity (FC), particularly concerning network formation, interaction, and dissolution over time, remains largely unexplored in AD.ObjectiveThis study aims to explore the effect of APOE ε4 on dynamic FC (dFC) variability and cognitive performance in AD.MethodsWe analyzed the dFC of AD patients, comparing APOE ε4 carriers (n = 33) with non-carriers (n = 41). The whole-brain dFC was assessed by calculating dynamic fractional amplitude of low-frequency fluctuations (dfALFF) and dynamic regional homogeneity (dReHo). To further explore the relationship between cognitive function and dFC in AD patients, we conducted a correlation analysis. Mediation analysis was also performed to determine whether dFC mediates the link between the APOE ε4 and cognitive decline in AD patients.ResultsAD patients carrying the APOE ε4 exhibited more severe cognitive impairment, along with reduced dReHo and dfALFF in both the left and right posterior cerebellar lobes. In these carriers, the dFC analysis showed lower dFC between the left posterior cerebellar lobe and the left middle temporal gyrus, which was positively correlated with executive function and information processing speed. Additionally, mediation analysis indicated that APOE ε4 influences dFC in this brain region, contributing to executive dysfunction in AD.ConclusionsThese findings offer preliminary evidence that APOE ε4 modulates fluctuating communication within the cerebellar lobe and the dFC between the cerebellar lobe and the temporal gyrus in AD.
Collapse
Affiliation(s)
- ChengBing Gong
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - WenTing Song
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - ZhengYang Zhu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Dan Yang
- Department of Neurology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| | - Xiang Zhao
- State Key Laboratory of Neurology and Oncology Drug Development, Jiangsu Simcere Diagnostics, Nanjing, China
- Simcere Medical Laboratory Science, Nanjing, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China
| | - Hui Zhao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China
| |
Collapse
|
2
|
Meyer-Acosta KK, Diaz-Guerra E, Varma P, Aruk A, Mirsadeghi S, Perez AM, Rafati Y, Hosseini A, Nieto-Estevez V, Giugliano M, Navara C, Hsieh J. APOE4 impacts cortical neurodevelopment and alters network formation in human brain organoids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.617044. [PMID: 39416105 PMCID: PMC11482793 DOI: 10.1101/2024.10.07.617044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Apolipoprotein E4 ( APOE4 ) is the leading genetic risk factor for Alzheimer's disease. While most studies examine the role of APOE4 in aging, imaging, and cognitive assessments reveal that APOE4 influences brain structure and function as early as infancy. Here, we examined human-relevant cellular phenotypes across neurodevelopment using induced pluripotent stem cell (iPSC) derived cortical and ganglionic eminence organoids (COs and GEOs). In COs, we showed that APOE4 decreased BRN2+ and SATB2+ cortical neurons, increased astrocytes and outer radial glia, and was associated with increased cell death and dysregulated GABA-related gene expression. In GEOs, APOE4 accelerated maturation of neural progenitors and neurons. Multi-electrode array recordings in assembloids revealed that APOE4 disrupted network formation and altered response to GABA, resulting in heightened excitability and synchronicity. Together, our data provides new insights into how APOE4 may influence cortical neurodevelopmental processes and network formation in the human brain.
Collapse
|
3
|
Mochel F. What can pediatricians learn from adult inherited metabolic diseases? J Inherit Metab Dis 2024; 47:876-884. [PMID: 38520225 DOI: 10.1002/jimd.12729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 03/02/2024] [Accepted: 03/05/2024] [Indexed: 03/25/2024]
Abstract
The field of inherited metabolic diseases (IMD) has initially emerged and developed over decades in pediatric departments. Still, today, about 50% of patients with IMD are adults, and adult metabolic medicine (AMM) is getting more structured at national and international levels. There are several domains in which pediatricians can learn from AMM. First, long-term evolution of IMD patients, especially those treated since childhood, is critical to determine nutritional and neuropsychiatric outcomes in adults so that these outcomes can be better monitored, and patient care adjusted as much as possible from childhood. Conversely, the observation of attenuated phenotypes in adults of IMD known to present with severe phenotypes in children calls for caution in the development of newborn screening programs and, more largely, in the interpretation of next-generation sequencing data. Third, it is important for pediatricians to be familiar with adult-onset IMD as they expand our understanding of metabolism, including in children, such as oxysterols and glycogen metabolism. Last, the identification of common molecular and cellular mechanisms in neurodevelopment and neurodegeneration opens the way to synergistic therapeutic developments that will benefit both fields of pediatric and adult medicine. Overall, these observations underline the need of strong interdisciplinarity between pediatricians and adult specialists for the diagnosis and the treatment of IMD well beyond the issues of patient transition from pediatric to adult medicine.
Collapse
Affiliation(s)
- Fanny Mochel
- AP-HP, Pitié-Salpêtrière University Hospital, Department of Medical Genetics, Reference Centers for Adult Neurometabolic Diseases and Adult Leukodystrophies, Paris, France
- INSERM U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Paris Brain Institute, ICM, Paris, France
| |
Collapse
|
4
|
Pardej SK, Casnar CL, Yund BD, Klein-Tasman BP. An evaluation of computerized attention and executive function measures for use with school age children with neurofibromatosis type 1. Child Neuropsychol 2024; 30:938-953. [PMID: 38214530 DOI: 10.1080/09297049.2024.2302634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 12/21/2023] [Indexed: 01/13/2024]
Abstract
The present study investigated the performance of children with neurofibromatosis type 1 on computerized assessments of attention and executive function. Relations to ADHD symptomatology were also examined. Participants included 37 children (20 male) with NF1 (9-13 years; Mage = 11.02). Participants completed the NIH Toolbox Dimensional Change Card Sort, List Sort Working Memory (LSWM), and Flanker tasks, as well as Cogstate Identification and One Back tests. ADHD symptomatology was assessed using the K-SADS. Average performance was significantly different from the normative mean on every measure, except LSWM. The NIH Toolbox Flanker and Cogstate Identification tasks detected the highest proportion of participants with at least mild difficulty, and the Cogstate Identification task detected the highest proportion of participants with severe difficulty. Analyses revealed significant relations with ADHD symptomatology for two NIH toolbox tasks. The various computerized measures of attention and executive function offer different information when working with school age children with NF1. The NIH Flanker may offer the most room for change and offers face validity, which may be beneficial for clinical trials research. However, the LSWM shows most support for relations with behavioral indicators of attention and executive challenges.
Collapse
Affiliation(s)
- Sara K Pardej
- Department of Psychology, University of Wisconsin - Milwaukee, Milwaukee, WI, USA
| | | | - Brianna D Yund
- Division of Clinical Behavioral Neuroscience, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | | |
Collapse
|
5
|
Lima-Cooper G, Ouma BJ, Datta D, Bond C, Soto AA, Conroy AL, Park GS, Bangirana P, Joloba ML, Opoka RO, Idro R, John CC. Apolipoprotein-E4: risk of severe malaria and mortality and cognitive impairment in pediatric cerebral malaria. Pediatr Res 2024; 96:89-96. [PMID: 38007518 PMCID: PMC12009649 DOI: 10.1038/s41390-023-02912-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 10/21/2023] [Accepted: 11/02/2023] [Indexed: 11/27/2023]
Abstract
BACKGROUND The relationship of apolipoprotein-E4 (APOE4) to mortality and cognition after severe malaria in children is unknown. METHODS APOE genotyping was performed in children with cerebral malaria (CM, n = 261), severe malarial anemia (SMA, n = 224) and community children (CC, n = 213). Cognition was assessed over 2-year follow-up. RESULTS A greater proportion of children with CM or SMA than CC had APOE4 (n = 162, 31.0%; n = 142, 31.7%; n = 103, 24.2%, respectively, p = 0.02), but no difference was seen in APOE3 (n = 310, 59.4%; n = 267, 59.6%; n = 282, 66.2%, respectively, p = 0.06), or APOE2 (n = 50, 9.6%; n = 39, 8.7%; and n = 41, 9.6%, respectively, p = 0.87). APOE4 was associated with increased mortality in CM (odds ratio, 2.28; 95% CI, 1.01, 5.11). However, APOE4 was associated with better long-term cognition (ß, 0.55; 95% CI, 0.04, 1.07, p = 0.04) and attention (ß 0.78; 95% CI, 0.26, 1.30, p = 0.004) in children with CM < 5 years old, but worse attention (ß, -0.90; 95% CI, -1.69, -0.10, p = 0.03) in children with CM ≥ 5 years old. Among children with CM, risk of post-discharge malaria was increased with APOE4 and decreased with APOE3. CONCLUSIONS APOE4 is associated with higher risk of CM or SMA and mortality in children with CM, but better long-term cognition in CM survivors <5 years of age.
Collapse
Affiliation(s)
- Giselle Lima-Cooper
- Department of Pediatrics, Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Benson J Ouma
- Department of Medical Microbiology, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Dibyadyuti Datta
- Department of Pediatrics, Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Caitlin Bond
- Department of Pediatrics, Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Alejandro A Soto
- Department of Pediatrics, Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrea L Conroy
- Department of Pediatrics, Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Gregory S Park
- Office of the Vice President for Research, University of Minnesota, Minneapolis, MN, USA
| | - Paul Bangirana
- Department of Psychiatry, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Moses L Joloba
- Department of Medical Microbiology, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Robert O Opoka
- Department of Pediatrics and Child Health, Makerere University, Kampala, Uganda
| | - Richard Idro
- Department of Pediatrics and Child Health, Makerere University, Kampala, Uganda
- Nuffield Department of Medicine, Centre of Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Chandy C John
- Department of Pediatrics, Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
6
|
Ogonowski NS, García-Marín LM, Fernando AS, Flores-Ocampo V, Rentería ME. Impact of genetic predisposition to late-onset neurodegenerative diseases on early life outcomes and brain structure. Transl Psychiatry 2024; 14:185. [PMID: 38605018 PMCID: PMC11009228 DOI: 10.1038/s41398-024-02898-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 03/27/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024] Open
Abstract
Most patients with late-onset neurodegenerative diseases such as Alzheimer's and Parkinson's have a complex aetiology resulting from numerous genetic risk variants of small effects located across the genome, environmental factors, and the interaction between genes and environment. Over the last decade, genome-wide association studies (GWAS) and post-GWAS analyses have shed light on the polygenic architecture of these diseases, enabling polygenic risk scores (PRS) to estimate an individual's relative genetic liability for presenting with the disease. PRS can screen and stratify individuals based on their genetic risk, potentially years or even decades before the onset of clinical symptoms. An emerging body of evidence from various research studies suggests that genetic susceptibility to late-onset neurodegenerative diseases might impact early life outcomes, including cognitive function, brain structure and function, and behaviour. This article summarises recent findings exploring the potential impact of genetic susceptibility to neurodegenerative diseases on early life outcomes. A better understanding of the impact of genetic susceptibility to neurodegenerative diseases early in life could be valuable in disease screening, detection, and prevention and in informing treatment strategies before significant neural damage has occurred. However, ongoing studies have limitations. Overall, our review found several studies focused on APOE haplotypes and Alzheimer's risk, but a limited number of studies leveraging polygenic risk scores or focused on genetic susceptibility to other late-onset conditions.
Collapse
Affiliation(s)
- Natalia S Ogonowski
- Mental Health & Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Luis M García-Marín
- Mental Health & Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Amali S Fernando
- Mental Health & Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Victor Flores-Ocampo
- Mental Health & Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Miguel E Rentería
- Mental Health & Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
7
|
Heise V, Offer A, Whiteley W, Mackay CE, Armitage JM, Parish S. A comprehensive analysis of APOE genotype effects on human brain structure in the UK Biobank. Transl Psychiatry 2024; 14:143. [PMID: 38472178 PMCID: PMC10933274 DOI: 10.1038/s41398-024-02848-5] [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: 07/27/2023] [Revised: 02/14/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Alzheimer's disease (AD) risk is increased in carriers of the apolipoprotein E (APOE) ε4 allele and decreased in ε2 allele carriers compared with the ε3ε3 genotype. The aim of this study was to determine whether: the APOE genotype affects brain grey (GM) or white matter (WM) structure; and if differences exist, the age when they become apparent and whether there are differential effects by sex. We used cross-sectional magnetic resonance imaging data from ~43,000 (28,494 after pre-processing) white British cognitively healthy participants (7,446 APOE ε4 carriers) aged 45-80 years from the UK Biobank cohort and investigated image-derived phenotypes (IDPs). We observed no statistically significant effects of APOE genotype on GM structure volumes or median T2* in subcortical structures, a measure related to iron content. The volume of white matter hyperintensities differed significantly between APOE genotype groups with higher volumes in APOE ε4ε4 (effect size 0.14 standard deviations [SD]) and ε3ε4 carriers (effect size 0.04 SD) but no differences in ε2 carriers compared with ε3ε3 carriers. WM integrity measures in the dorsal (mean diffusivity [MD]) and ventral cingulum (MD and intracellular volume fraction), posterior thalamic radiation (MD and isotropic volume fraction) and sagittal stratum (MD) indicated lower integrity in APOE ε4ε4 carriers (effect sizes around 0.2-0.3 SD) and ε3ε4 (effect sizes around 0.05 SD) carriers but no differences in ε2 carriers compared with the APOE ε3ε3 genotype. Effects did not differ between men and women. APOE ε4 homozygotes had lower WM integrity specifically at older ages with a steeper decline of WM integrity from the age of 60 that corresponds to around 5 years greater "brain age". APOE genotype affects various white matters measures, which might be indicative of preclinical AD processes. This hypothesis can be assessed in future when clinical outcomes become available.
Collapse
Affiliation(s)
- Verena Heise
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Alison Offer
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - William Whiteley
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Clare E Mackay
- Department of Psychiatry, University of Oxford, Oxford, UK
- Wellcome Centre for Integrative Neuroimaging, Oxford Centre for Human Brain Activity, University of Oxford, Oxford, UK
| | - Jane M Armitage
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Sarah Parish
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK.
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK.
| |
Collapse
|
8
|
Kuhn HG, Skau S, Nyberg J. A lifetime perspective on risk factors for cognitive decline with a special focus on early events. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2024; 6:100217. [PMID: 39071743 PMCID: PMC11273094 DOI: 10.1016/j.cccb.2024.100217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 07/30/2024]
Abstract
Both Alzheimer's disease and vascular dementia are the result of disease processes that typically develop over several decades. Population studies have estimated that more than half of the risk for dementia is preventable or at least modifiable through behavioral adaptations. The association between these lifestyle factors and the risk of dementia is most evident for exposure in midlife. However, habits formed in middle age often reflect a lifetime of behavior patterns and living conditions. Therefore, individuals who, for example, are able to maintain healthy diets and regular exercise during their middle years are likely to benefit from these cognition-protective habits they have practiced throughout their lives. For numerous adult diseases, significant risks can often be traced back to early childhood. Suboptimal conditions during the perinatal period, childhood and adolescence can increase the risk of adult diseases, including stroke, heart disease, insulin resistance, hypertension and dementia. This review aims at summarizing some of the evidence for dementia risks from a life-time perspective with the goal of raising awareness for early dementia prevention and successful aging.
Collapse
Affiliation(s)
- H. Georg Kuhn
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Simon Skau
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
- Department of Pedagogical, Curricular and Professional Studies, University of Gothenburg, Gothenburg, Sweden
| | - Jenny Nyberg
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
9
|
Wang HA, Liang HJ, Ernst TM, Nakama H, Cunningham E, Chang L. Independent and combined effects of methamphetamine use disorders and APOEε4 allele on cognitive performance and brain morphometry. Addiction 2023; 118:2384-2396. [PMID: 37563863 PMCID: PMC10840926 DOI: 10.1111/add.16309] [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: 12/18/2022] [Accepted: 06/19/2023] [Indexed: 08/12/2023]
Abstract
AIMS Prior studies showed that methamphetamine (METH) users had greater than normal age-related brain atrophy; whether having the apolipoprotein E (APOE)-ε4 allele may be a contributory factor has not been evaluated. We aimed to determine the independent and combined effects of chronic heavy METH use and having at least one copy of the APOE-ε4 allele (APOE-ε4+) on brain morphometry and cognition, especially in relation to aging. METHODS We compared brain morphometry and cognitive performance in 77 individuals with chronic heavy METH use (26 APOE-ε4+, 51 APOE-ε4-) and 226 Non-METH users (66 APOE-ε4+, 160 APOE-ε4-), using a 2 × 2 design (two-way analysis of co-variance). Vertex-wise cortical volumes, thickness and seven subcortical volumes, were automatically measured using FreeSurfer. Linear regression between regional brain measures, and cognitive scores that showed group differences were evaluated. Group differences in age-related decline in brain and cognitive measures were also explored. RESULTS Regardless of APOE-ε4 genotype, METH users had lower Motor Z-scores (P = 0.005), thinner right lateral-orbitofrontal cortices (P < 0.001), smaller left pars-triangularis gyrus volumes (P = 0.004), but larger pallida, hippocampi and amygdalae (P = 0.004-0.006) than nonusers. Across groups, APOE-ε4+ METH users had the smallest volumes of superior frontal cortical gyri bilaterally, and of the smallest volume in left rostral-middle frontal gyri (all P-values <0.001). Smaller right superior-frontal gyrus predicted poorer motor function only in APOE-ε4+ participants (interaction-P < 0.001). Cortical volumes and thickness declined with age similarly across all participants; however, APOE-ε4-carriers showed thinner right inferior parietal cortices than noncarriers at younger age (interaction-P < 0.001). CONCLUSIONS Chronic heavy use and having at least one copy of the APOE-ε4 allele may have synergistic effects on brain atrophy, particularly in frontal cortices, which may contribute to their poorer cognitive function. However, the enlarged subcortical volumes in METH users replicated prior studies, and are likely due to METH-mediated neuroinflammation.
Collapse
Affiliation(s)
- Hannah A. Wang
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Hua-Jun Liang
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Thomas M. Ernst
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Eric Cunningham
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Linda Chang
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
10
|
Nichols E, Brickman AM, Casaletto KB, Dams-O’Connor K, George KM, Kumar RG, Palta P, Rabin JS, Satizabal CL, Schneider J, Pa J, La Joie R. AD and non-AD mediators of the pathway between the APOE genotype and cognition. Alzheimers Dement 2023; 19:2508-2519. [PMID: 36516004 PMCID: PMC10264550 DOI: 10.1002/alz.12885] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 12/15/2022]
Abstract
INTRODUCTION The apolipoprotein E (APOE) genotype is a driver of cognitive decline and dementia. We used causal mediation methods to characterize pathways linking the APOE genotype to late-life cognition through Alzheimer's disease (AD) and non-AD neuropathologies. METHODS We analyzed autopsy data from 1671 individuals from the Religious Orders Study, Memory and Aging Project, and Minority Aging Research Study (ROS/MAP/MARS) studies with cognitive assessment within 5 years of death and autopsy measures of AD (amyloid beta (Aβ), neurofibrillary tangles), vascular (athero/arteriolo-sclerosis, micro-infarcts/macro-infarcts), and non-AD neurodegenerative neuropathologies (TAR DNA protein 43 [TDP-43], Lewy bodies, amyloid angiopathy, hippocampal sclerosis). RESULTS The detrimental effect of APOE ε4 on cognition was mediated by summary measures of AD and non-AD neurodegenerative neuropathologies but not vascular neuropathologies; effects were strongest in individuals with dementia. The protective effect of APOE ε2 was partly mediated by AD neuropathology and stronger in women than in men. DISCUSSION The APOE genotype influences cognition and dementia through multiple neuropathological pathways, with implications for different therapeutic strategies targeting people at increased risk for dementia. HIGHLIGHTS Both apolipoprotein E (APOE) ε2 and APOE ε4 effects on late-life cognition are mediated by AD neuropathology. The estimated mediated effects of most measures of AD neuropathology were similar. Non-Alzheimer's disease (AD) neurodegenerative pathologies mediate the effect of ε4 independently from AD. Non-AD vascular pathologies did not mediate the effect of the APOE genotype on cognition. The protective effect of APOE ε2 on cognition was stronger in women.
Collapse
Affiliation(s)
- Emma Nichols
- Department of Epidemiology, Johns Hopkins Bloomberg School
of Public Health, Baltimore, MD, USA
| | - Adam M. Brickman
- Taub Institute for Research on Alzheimer’s Disease
and the Aging Brain, Department of Neurology, College of Physicians and Surgeons,
Columbia University, New York, NY, USA
| | - Kaitlin B. Casaletto
- Memory and Aging Center, Department of Neurology, Weill
Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Kristen Dams-O’Connor
- Department of Rehabilitation and Human Performance, Icahn
School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Icahn School of Medicine at Mount
Sinai, New York, NY, USA
| | - Kristen M. George
- Department of Public Health Sciences, University of
California Davis School of Medicine, Davis, CA, USA
| | - Raj G. Kumar
- Department of Rehabilitation and Human Performance, Icahn
School of Medicine at Mount Sinai, New York, NY, USA
| | - Priya Palta
- Departments of Medicine and Epidemiology, Columbia
University Irving Medical Center, New York, NY, USA
| | - Jennifer S. Rabin
- Division of Neurology, Department of Medicine, Sunnybrook
Health Sciences Centre, University of Toronto, Canada
- Harquail Centre for Neuromodulation, Hurvitz Brain
Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Rehabilitation Sciences Institute, University of
Toronto, Canada
| | - Claudia L. Satizabal
- Department of Population Health Science and Biggs
Institute for Alzheimer’s and Neurodegenerative Diseases, UT Health San
Antonio, San Antonio, TX, USA
- Department of Neurology, Boston University School of
Medicine, Boston, MA, USA
| | - Julie Schneider
- Rush Alzheimer’s Disease Center, Chicago, IL,
USA
- Rush University Medical Center, Chicago, IL, USA
| | - Judy Pa
- Department of Neuroscience, University of California San
Diego, San Diego, CA, USA
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill
Institute for Neurosciences, University of California, San Francisco, CA, USA
| |
Collapse
|
11
|
Kline C, Stoller S, Byer L, Samuel D, Lupo JM, Morrison MA, Rauschecker AM, Nedelec P, Faig W, Dubal DB, Fullerton HJ, Mueller S. An Integrated Analysis of Clinical, Genomic, and Imaging Features Reveals Predictors of Neurocognitive Outcomes in a Longitudinal Cohort of Pediatric Cancer Survivors, Enriched with CNS Tumors (Rad ART Pro). Front Oncol 2022; 12:874317. [PMID: 35814456 PMCID: PMC9259981 DOI: 10.3389/fonc.2022.874317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
Background Neurocognitive deficits in pediatric cancer survivors occur frequently; however, individual outcomes are unpredictable. We investigate clinical, genetic, and imaging predictors of neurocognition in pediatric cancer survivors, with a focus on survivors of central nervous system (CNS) tumors exposed to radiation. Methods One hundred eighteen patients with benign or malignant cancers (median diagnosis age: 7; 32% embryonal CNS tumors) were selected from an existing multi-institutional cohort (RadART Pro) if they had: 1) neurocognitive evaluation; 2) available DNA; 3) standard imaging. Utilizing RadART Pro, we collected clinical history, genomic sequencing, CNS imaging, and neurocognitive outcomes. We performed single nucleotide polymorphism (SNP) genotyping for candidate genes associated with neurocognition: COMT, BDNF, KIBRA, APOE, KLOTHO. Longitudinal neurocognitive testing were performed using validated computer-based CogState batteries. The imaging cohort was made of patients with available iron-sensitive (n = 28) and/or T2 FLAIR (n = 41) sequences. Cerebral microbleeds (CMB) were identified using a semi-automated algorithm. Volume of T2 FLAIR white matter lesions (WML) was measured using an automated method based on a convolutional neural network. Summary statistics were performed for patient characteristics, neurocognitive assessments, and imaging. Linear mixed effects and hierarchical models assessed patient characteristics and SNP relationship with neurocognition over time. Nested case-control analysis was performed to compare candidate gene carriers to non-carriers. Results CMB presence at baseline correlated with worse performance in 3 of 7 domains, including executive function. Higher baseline WML volumes correlated with worse performance in executive function and verbal learning. No candidate gene reliably predicted neurocognitive outcomes; however, APOE ϵ4 carriers trended toward worse neurocognitive function over time compared to other candidate genes and carried the highest odds of low neurocognitive performance across all domains (odds ratio 2.85, P=0.002). Hydrocephalus and seizures at diagnosis were the clinical characteristics most frequently associated with worse performance in neurocognitive domains (5 of 7 domains). Overall, executive function and verbal learning were the most frequently negatively impacted neurocognitive domains. Conclusion Presence of CMB, APOE ϵ4 carrier status, hydrocephalus, and seizures correlate with worse neurocognitive outcomes in pediatric cancer survivors, enriched with CNS tumors exposed to radiation. Ongoing research is underway to verify trends in larger cohorts.
Collapse
Affiliation(s)
- Cassie Kline
- Division of Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Division of Child Neurology, Department of Neurology, University of California, San Francisco, United States
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| | - Schuyler Stoller
- Division of Child Neurology, Department of Neurology, University of California, San Francisco, United States
| | - Lennox Byer
- UCSF School of Medicine, University of California, San Francisco, United States
| | - David Samuel
- Division of Pediatric Hematology/Oncology, Valley Children’s Hospital, Madera, CA, United States
| | - Janine M. Lupo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, United States
| | - Melanie A. Morrison
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, United States
| | - Andreas M. Rauschecker
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, United States
| | - Pierre Nedelec
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, United States
| | - Walter Faig
- Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Dena B. Dubal
- Department of Neurology, University of California, San Francisco, CA, United States
| | - Heather J. Fullerton
- Division of Child Neurology, Department of Neurology, University of California, San Francisco, United States
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| | - Sabine Mueller
- Division of Child Neurology, Department of Neurology, University of California, San Francisco, United States
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
- Department of Neurological Surgery, University of California, San Francisco, CA, United States
- *Correspondence: Sabine Mueller,
| |
Collapse
|
12
|
Leitner DF, William C, Faustin A, Askenazi M, Kanshin E, Snuderl M, McGuone D, Wisniewski T, Ueberheide B, Gould L, Devinsky O. Proteomic differences in hippocampus and cortex of sudden unexplained death in childhood. Acta Neuropathol 2022; 143:585-599. [PMID: 35333953 PMCID: PMC8953962 DOI: 10.1007/s00401-022-02414-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 11/01/2022]
Abstract
Sudden unexplained death in childhood (SUDC) is death of a child over 1 year of age that is unexplained after review of clinical history, circumstances of death, and complete autopsy with ancillary testing. Multiple etiologies may cause SUDC. SUDC and sudden unexpected death in epilepsy (SUDEP) share clinical and pathological features, suggesting some similarities in mechanism of death and possible abnormalities in hippocampus and cortex. To identify molecular signaling pathways, we performed label-free quantitative mass spectrometry on microdissected frontal cortex, hippocampal dentate gyrus (DG), and cornu ammonis (CA1-3) in SUDC (n = 19) and pediatric control cases (n = 19) with an explained cause of death. At a 5% false discovery rate (FDR), we found differential expression of 660 proteins in frontal cortex, 170 in DG, and 57 in CA1-3. Pathway analysis of altered proteins identified top signaling pathways associated with activated oxidative phosphorylation (p = 6.3 × 10-15, z = 4.08) and inhibited EIF2 signaling (p = 2.0 × 10-21, z = - 2.56) in frontal cortex, and activated acute phase response in DG (p = 8.5 × 10-6, z = 2.65) and CA1-3 (p = 4.7 × 10-6, z = 2.00). Weighted gene correlation network analysis (WGCNA) of clinical history indicated that SUDC-positive post-mortem virology (n = 4/17) had the most significant module in each brain region, with the top most significant associated with decreased mRNA metabolic processes (p = 2.8 × 10-5) in frontal cortex. Additional modules were associated with clinical history, including fever within 24 h of death (top: increased mitochondrial fission in DG, p = 1.8 × 10-3) and febrile seizure history (top: decreased small molecule metabolic processes in frontal cortex, p = 8.8 × 10-5) in all brain regions, neuropathological hippocampal findings in the DG (top: decreased focal adhesion, p = 1.9 × 10-3). Overall, cortical and hippocampal protein changes were present in SUDC cases and some correlated with clinical features. Our studies support that proteomic studies of SUDC cohorts can advance our understanding of the pathogenesis of these tragedies and may inform the development of preventive strategies.
Collapse
|
13
|
Hol HR, Flak MM, Chang L, Løhaugen GCC, Bjuland KJ, Rimol LM, Engvig A, Skranes J, Ernst T, Madsen BO, Hernes SS. Cortical Thickness Changes After Computerized Working Memory Training in Patients With Mild Cognitive Impairment. Front Aging Neurosci 2022; 14:796110. [PMID: 35444526 PMCID: PMC9014119 DOI: 10.3389/fnagi.2022.796110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
Background Adaptive computerized working memory (WM) training has shown favorable effects on cerebral cortical thickness as compared to non-adaptive training in healthy individuals. However, knowledge of WM training-related morphological changes in mild cognitive impairment (MCI) is limited. Objective The primary objective of this double-blind randomized study was to investigate differences in longitudinal cortical thickness trajectories after adaptive and non-adaptive WM training in patients with MCI. We also investigated the genotype effects on cortical thickness trajectories after WM training combining these two training groups using longitudinal structural magnetic resonance imaging (MRI) analysis in Freesurfer. Method Magnetic resonance imaging acquisition at 1.5 T were performed at baseline, and after four- and 16-weeks post training. A total of 81 individuals with MCI accepted invitations to undergo 25 training sessions over 5 weeks. Longitudinal Linear Mixed effect models investigated the effect of adaptive vs. non-adaptive WM training. The LME model was fitted for each location (vertex). On all statistical analyzes, a threshold was applied to yield an expected false discovery rate (FDR) of 5%. A secondary LME model investigated the effects of LMX1A and APOE-ε4 on cortical thickness trajectories after WM training. Results A total of 62 participants/patients completed the 25 training sessions. Structural MRI showed no group difference between the two training regimes in our MCI patients, contrary to previous reports in cognitively healthy adults. No significant structural cortical changes were found after training, regardless of training type, across all participants. However, LMX1A-AA carriers displayed increased cortical thickness trajectories or lack of decrease in two regions post-training compared to those with LMX1A-GG/GA. No training or training type effects were found in relation to the APOE-ε4 gene variants. Conclusion The MCI patients in our study, did not have improved cortical thickness after WM training with either adaptive or non-adaptive training. These results were derived from a heterogeneous population of MCI participants. The lack of changes in the cortical thickness trajectory after WM training may also suggest the lack of atrophy during this follow-up period. Our promising results of increased cortical thickness trajectory, suggesting greater neuroplasticity, in those with LMX1A-AA genotype need to be validated in future trials.
Collapse
Affiliation(s)
- Haakon R. Hol
- Department of Radiology, Sørlandet Hospital, Arendal, Norway
- Department of Radiology, Oslo University Hospital, Oslo, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
- *Correspondence: Haakon R. Hol,
| | | | - Linda Chang
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | | | - Knut Jørgen Bjuland
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Lars M. Rimol
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Andreas Engvig
- Department of Medicine, Diakonhjemmet Hospital, Oslo, Norway
| | - Jon Skranes
- Department of Pediatrics, Sørlandet Hospital, Arendal, Norway
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Thomas Ernst
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Bengt-Ove Madsen
- Department of Geriatric and Internal Medicine, Sørlandet Hospital, Arendal, Norway
| | - Susanne S. Hernes
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Geriatric and Internal Medicine, Sørlandet Hospital, Arendal, Norway
| |
Collapse
|
14
|
Bilingualism is a long-term cognitively challenging experience that modulates metabolite concentrations in the healthy brain. Sci Rep 2021; 11:7090. [PMID: 33782462 PMCID: PMC8007713 DOI: 10.1038/s41598-021-86443-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/16/2021] [Indexed: 02/01/2023] Open
Abstract
Cognitively demanding experiences, including complex skill acquisition and processing, have been shown to induce brain adaptations, at least at the macroscopic level, e.g. on brain volume and/or functional connectivity. However, the neurobiological bases of these adaptations, including at the cellular level, are unclear and understudied. Here we use bilingualism as a case study to investigate the metabolic correlates of experience-based brain adaptations. We employ Magnetic Resonance Spectroscopy to measure metabolite concentrations in the basal ganglia, a region critical to language control which is reshaped by bilingualism. Our results show increased myo-Inositol and decreased N-acetyl aspartate concentrations in bilinguals compared to monolinguals. Both metabolites are linked to synaptic pruning, a process underlying experience-based brain restructuring. Interestingly, both concentrations correlate with relative amount of bilingual engagement. This suggests that degree of long-term cognitive experiences matters at the level of metabolic concentrations, which might accompany, if not drive, macroscopic brain adaptations.
Collapse
|
15
|
Hernes SS, Flak MM, Løhaugen GCC, Skranes J, Hol HR, Madsen BO, Knapskog AB, Engvig A, Pripp A, Ulstein I, Lona T, Zhang X, Chang L. Working Memory Training in Amnestic and Non-amnestic Patients With Mild Cognitive Impairment: Preliminary Findings From Genotype Variants on Training Effects. Front Aging Neurosci 2021; 13:624253. [PMID: 33658917 PMCID: PMC7917210 DOI: 10.3389/fnagi.2021.624253] [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: 10/30/2020] [Accepted: 01/12/2021] [Indexed: 11/13/2022] Open
Abstract
Working memory training (WMT) effects may be modulated by mild cognitive impairment (MCI) subtypes, and variations in APOE-epsilon (APOE-ε) and LMX1A genotypes. Sixty-one individuals (41 men/20 women, mean age 66 years) diagnosed with MCI (31 amnestic/30 non-amnestic) and genotyped for APOE-ε and LMX1A completed 4 weeks/20-25 sessions of WMT. Cognitive functions were assessed before, 4 weeks and 16 weeks after WMT. Except for Processing Speed, the non-amnestic MCI group (naMCI) outperformed the amnestic MCI (aMCI) group in all cognitive domains across all time-points. At 4 weeks, working memory function improved in both groups (p < 0.0001), but at 16 weeks the effects only remained in the naMCI group. Better performance was found after training for the naMCI patients with LMX1A-AA genotype and for the APOE-ε4 carriers. Only the naMCI-APOE-ε4 group showed improved Executive Function at 16 weeks. WMT improved working memory and some non-trained cognitive functions in individuals with MCI. The naMCI group had greater training gain than aMCI group, especially in those with LMX1A-AA genotype and among APOE-ε4-carriers. Further research with larger sample sizes for the subgroups and longer follow-up evaluations is warranted.
Collapse
Affiliation(s)
- Susanne S Hernes
- Department of Geriatric and Internal Medicine, Sørlandet Hospital, Arendal, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Marianne M Flak
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Pediatrics, Sørlandet Hospital HF, Arendal, Norway
| | - Gro C C Løhaugen
- Department of Pediatrics, Sørlandet Hospital HF, Arendal, Norway
| | - Jon Skranes
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Pediatrics, Sørlandet Hospital HF, Arendal, Norway
| | - Haakon R Hol
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Radiology, Sørlandet Hospital HF, Arendal, Norway
| | - Bengt-Ove Madsen
- Department of Geriatric and Internal Medicine, Sørlandet Hospital, Arendal, Norway
| | - Anne-Brita Knapskog
- Department of Geriatric Medicine, The Memory Clinic, Oslo University Hospital, Oslo, Norway
| | - Andreas Engvig
- Department of Medicine, Diakonhjemmet Hospital, Oslo, Norway
| | - Are Pripp
- Oslo Centre of Biostatistics and Epidemiology Research Support Services, Oslo University Hospital, Oslo, Norway
| | - Ingun Ulstein
- Department of Geriatric Medicine, The Memory Clinic, Oslo University Hospital, Oslo, Norway
| | - Trine Lona
- Department of Psychiatry, Age Psychiatry, The Hospital of Telemark, Skien, Norway
| | - Xin Zhang
- Department of Diagnostic Radiology and Nuclear Medicine, and Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Linda Chang
- Department of Diagnostic Radiology and Nuclear Medicine, and Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Medicine, John A. Burns School of Medicine, The University of Hawai'i at Mānoa, Honolulu, HI, United States
| |
Collapse
|
16
|
Li Z, Shue F, Zhao N, Shinohara M, Bu G. APOE2: protective mechanism and therapeutic implications for Alzheimer's disease. Mol Neurodegener 2020; 15:63. [PMID: 33148290 PMCID: PMC7640652 DOI: 10.1186/s13024-020-00413-4] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 10/17/2020] [Indexed: 02/06/2023] Open
Abstract
Investigations of apolipoprotein E (APOE) gene, the major genetic risk modifier for Alzheimer's disease (AD), have yielded significant insights into the pathogenic mechanism. Among the three common coding variants, APOE*ε4 increases, whereas APOE*ε2 decreases the risk of late-onset AD compared with APOE*ε3. Despite increased understanding of the detrimental effect of APOE*ε4, it remains unclear how APOE*ε2 confers protection against AD. Accumulating evidence suggests that APOE*ε2 protects against AD through both amyloid-β (Aβ)-dependent and independent mechanisms. In addition, APOE*ε2 has been identified as a longevity gene, suggesting a systemic effect of APOE*ε2 on the aging process. However, APOE*ε2 is not entirely benign; APOE*ε2 carriers exhibit increased risk of certain cerebrovascular diseases and neurological disorders. Here, we review evidence from both human and animal studies demonstrating the protective effect of APOE*ε2 against AD and propose a working model depicting potential underlying mechanisms. Finally, we discuss potential therapeutic strategies designed to leverage the protective effect of APOE2 to treat AD.
Collapse
Affiliation(s)
- Zonghua Li
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Francis Shue
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Neuroscience Graduate Program, Mayo Clinic, Jacksonville, FL, USA
| | - Na Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Mitsuru Shinohara
- Department of Aging Neurobiology, National Center for Geriatrics and Gerontology, 7-430 Morioka, Obu, Aichi, 474-8511, Japan.
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
- Neuroscience Graduate Program, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
17
|
The effect of bilingualism on brain development from early childhood to young adulthood. Brain Struct Funct 2020; 225:2131-2152. [PMID: 32691216 PMCID: PMC7473972 DOI: 10.1007/s00429-020-02115-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/09/2020] [Indexed: 11/30/2022]
Abstract
Bilingualism affects the structure of the brain in adults, as evidenced by experience-dependent grey and white matter changes in brain structures implicated in language learning, processing, and control. However, limited evidence exists on how bilingualism may influence brain development. We examined the developmental patterns of both grey and white matter structures in a cross-sectional study of a large sample (n = 711 for grey matter, n = 637 for white matter) of bilingual and monolingual participants, aged 3–21 years. Metrics of grey matter (thickness, volume, and surface area) and white matter (fractional anisotropy and mean diffusivity) were examined across 41 cortical and subcortical brain structures and 20 tracts, respectively. We used generalized additive modelling to analyze whether, how, and where the developmental trajectories of bilinguals and monolinguals might differ. Bilingual and monolingual participants manifested distinct developmental trajectories in both grey and white matter structures. As compared to monolinguals, bilinguals showed: (a) more grey matter (less developmental loss) starting during late childhood and adolescence, mainly in frontal and parietal regions (particularly in the inferior frontal gyrus pars opercularis, superior frontal cortex, inferior and superior parietal cortex, and precuneus); and (b) higher white matter integrity (greater developmental increase) starting during mid-late adolescence, specifically in striatal–inferior frontal fibers. The data suggest that there may be a developmental basis to the well-documented structural differences in the brain between bilingual and monolingual adults.
Collapse
|
18
|
Yassine HN, Finch CE. APOE Alleles and Diet in Brain Aging and Alzheimer's Disease. Front Aging Neurosci 2020; 12:150. [PMID: 32587511 PMCID: PMC7297981 DOI: 10.3389/fnagi.2020.00150] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 05/04/2020] [Indexed: 12/13/2022] Open
Abstract
The APOE gene alleles modify human aging and the response to the diet at many levels with diverse pleotropic effects from gut to brain. To understand the interactions of APOE isoforms and diet, we analyze how cellular trafficking of apoE proteins affects energy metabolism, the immune system, and reproduction. The age-accelerating APOE4 allele alters the endosomal trafficking of cell surface receptors that mediate lipid and glucose metabolism. The APOE4 allele is the ancestral human allele, joined by APOE3 and then APOE2 in the human species. Under conditions of high infection, uncertain food, and shorter life expectancy, APOE4 may be adaptive for reducing mortality. As humans transitioned into modern less-infectious environments and longer life spans, APOE4 increased risks of aging-related diseases, particularly impacting arteries and the brain. The association of APOE4 with glucose dysregulation and body weight promotes many aging-associated diseases. Additionally, the APOE gene locus interacts with adjacent genes on chromosome 19 in haplotypes that modify neurodegeneration and metabolism, for which we anticipate complex gene-environment interactions. We summarize how diet and Alzheimer's disease (AD) risk are altered by APOE genotype in both animal and human studies and identify gaps. Much remains obscure in how APOE alleles modify nutritional factors in human aging. Identifying risk variant haplotypes in the APOE gene complex will clarify homeostatic adaptive responses to environmental conditions.
Collapse
Affiliation(s)
- Hussein N. Yassine
- Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Caleb E. Finch
- Leonard Davis School of Gerontology and Dornsife College, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
19
|
Vinueza-Veloz MF, Martín-Román C, Robalino-Valdivieso MP, White T, Kushner SA, De Zeeuw CI. Genetic risk for Alzheimer disease in children: Evidence from early-life IQ and brain white-matter microstructure. GENES BRAIN AND BEHAVIOR 2020; 19:e12656. [PMID: 32383552 PMCID: PMC7507145 DOI: 10.1111/gbb.12656] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/01/2020] [Accepted: 04/17/2020] [Indexed: 01/21/2023]
Abstract
It remains unclear whether the genetic risk for late‐onset Alzheimer disease (AD) is linked to premorbid individual differences in general cognitive ability and brain structure. The objective of the present study was to determine whether the genetic risk of late‐onset AD is related to premorbid individual differences in intelligence quotient (IQ) and characteristics of the cerebral white‐matter in children. The study sample included children of the Generation R Study from Rotterdam, The Netherlands. IQ was measured using a well‐validated Dutch nonverbal IQ test (n = 1908) at ages 5 to 9 years. White‐matter microstructure was assessed by measuring fractional anisotropy (FA) of white‐matter tracts using diffusion tensor imaging (DTI) (n = 919) at ages 9 to 12 years. Genetic risk was quantified using three biologically defined genetic risk scores (GRSs) hypothesized to be related to the pathophysiology of late‐onset AD: immune response, cholesterol/lipid metabolism and endocytosis. Higher genetic risk for late‐onset AD that included genes associated with immune responsivity had a negative influence on cognition and cerebral white‐matter microstructure. For each unit increase in the immune response GRS, IQ decreased by 0.259 SD (95% CI [−0.500, −0.017]). For each unit increase in the immune response GRS, global FA decreased by 0.373 SD (95% CI [−0.721, −0.026]). Neither cholesterol/lipid metabolism nor endocytosis GRSs were associated with IQ or cerebral white‐matter microstructure. Our findings suggest that elevated genetic risk for late‐onset AD may in part be manifest during childhood neurodevelopment through alterations in immune responsivity.
Collapse
Affiliation(s)
- María Fernanda Vinueza-Veloz
- School of Medicine, Escuela Superior Politécnica de Chimborazo, Riobamba, Ecuador.,Department of Neuroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Carlos Martín-Román
- Leiden Institute for Advanced Computer Science, Leiden University, Leiden, The Netherlands
| | | | - Tonya White
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus MC, Rotterdam, The Netherlands.,Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Steven A Kushner
- Department of Psychiatry, Erasmus MC, Rotterdam, The Netherlands.,Department of Psychiatry, Columbia University, New York City, United States of America, United States of America
| | - Chris I De Zeeuw
- Department of Neuroscience, Erasmus MC, Rotterdam, The Netherlands.,Royal Netherlands Academy of Arts and Sciences, The Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| |
Collapse
|
20
|
Iacono D, Feltis GC. Impact of Apolipoprotein E gene polymorphism during normal and pathological conditions of the brain across the lifespan. Aging (Albany NY) 2020; 11:787-816. [PMID: 30677746 PMCID: PMC6366964 DOI: 10.18632/aging.101757] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 01/05/2019] [Indexed: 12/12/2022]
Abstract
The central nervous system (CNS) is the cellular substrate for the integration of complex, dynamic, constant, and simultaneous interactions among endogenous and exogenous stimuli across the entire human lifespan. Numerous studies on aging-related brain diseases show that some genes identified as risk factors for some of the most common neurodegenerative diseases - such as the allele 4 of APOE gene (APOE4) for Alzheimer's disease (AD) - have a much earlier neuro-anatomical and neuro-physiological impact. The impact of APOE polymorphism appears in fact to start as early as youth and early-adult life. Intriguingly, though, those same genes associated with aging-related brain diseases seem to influence different aspects of the brain functioning much earlier actually, that is, even from the neonatal periods and earlier. The APOE4, an allele classically associated with later-life neurodegenerative disorders as AD, seems in fact to exert a series of very early effects on phenomena of neuroplasticity and synaptogenesis that begin from the earliest periods of life such as the fetal ones.We reviewed some of the findings supporting the hypothesis that APOE polymorphism is an early modifier of various neurobiological aspects across the entire human lifespan - from the in-utero to the centenarian life - during both normal and pathological conditions of the brain.
Collapse
Affiliation(s)
- Diego Iacono
- Neuropathology Research, Biomedical Research Institute of New Jersey (BRInj), Cedar Knolls, NJ 07927, USA.,MidAtlantic Neonatology Associates (MANA), Morristown, NJ 07960, USA.,Atlantic Neuroscience Institute, Atlantic Health System (AHS), Overlook Medical Center, Summit, NJ 07901, USA
| | - Gloria C Feltis
- Neuropathology Research, Biomedical Research Institute of New Jersey (BRInj), Cedar Knolls, NJ 07927, USA
| |
Collapse
|
21
|
Cacciaglia R, Molinuevo JL, Falcón C, Arenaza-Urquijo EM, Sánchez-Benavides G, Brugulat-Serrat A, Blennow K, Zetterberg H, Gispert JD. APOE-ε4 Shapes the Cerebral Organization in Cognitively Intact Individuals as Reflected by Structural Gray Matter Networks. Cereb Cortex 2020; 30:4110-4120. [PMID: 32163130 PMCID: PMC7264689 DOI: 10.1093/cercor/bhaa034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/28/2020] [Accepted: 01/30/2020] [Indexed: 11/19/2022] Open
Abstract
Gray matter networks (GMn) provide essential information on the intrinsic organization of the brain and appear to be disrupted in Alzheimer’s disease (AD). Apolipoprotein E (APOE)-ε4 represents the major genetic risk factor for AD, yet the association between APOE-ε4 and GMn has remained unexplored. Here, we determine the impact of APOE-ε4 on GMn in a large sample of cognitively unimpaired individuals, which was enriched for the genetic risk of AD. We used independent component analysis to retrieve sources of structural covariance and analyzed APOE group differences within and between networks. Analyses were repeated in a subsample of amyloid-negative subjects. Compared with noncarriers and heterozygotes, APOE-ε4 homozygotes showed increased covariance in one network including primarily right-lateralized, parietal, inferior frontal, as well as inferior and middle temporal regions, which mirrored the formerly described AD-signature. This result was confirmed in a subsample of amyloid-negative individuals. APOE-ε4 carriers showed reduced covariance between two networks encompassing frontal and temporal regions, which constitute preferential target of amyloid deposition. Our data indicate that, in asymptomatic individuals, APOE-ε4 shapes the cerebral organization in a way that recapitulates focal morphometric alterations observed in AD patients, even in absence of amyloid pathology. This suggests that structural vulnerability in neuronal networks associated with APOE-ε4 may be an early event in AD pathogenesis, possibly upstream of amyloid deposition.
Collapse
Affiliation(s)
- Raffaele Cacciaglia
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, 08005 Barcelona, Spain.,Hospital del Mar Medical Research Institute (IMIM), 08005 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), 28089 Madrid, Spain
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, 08005 Barcelona, Spain.,Hospital del Mar Medical Research Institute (IMIM), 08005 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), 28089 Madrid, Spain.,Universitat Pompeu Fabra, 08002 Barcelona, Spain
| | - Carles Falcón
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, 08005 Barcelona, Spain.,Hospital del Mar Medical Research Institute (IMIM), 08005 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBERBBN), 28089 Madrid, Spain
| | - Eider M Arenaza-Urquijo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, 08005 Barcelona, Spain.,Hospital del Mar Medical Research Institute (IMIM), 08005 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), 28089 Madrid, Spain
| | - Gonzalo Sánchez-Benavides
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, 08005 Barcelona, Spain.,Hospital del Mar Medical Research Institute (IMIM), 08005 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), 28089 Madrid, Spain
| | - Anna Brugulat-Serrat
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, 08005 Barcelona, Spain.,Hospital del Mar Medical Research Institute (IMIM), 08005 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), 28089 Madrid, Spain.,Global Brain Health Institute, University of California San Francisco, San Francisco, CA 94115, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, 41390 Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 41390 Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, 41390 Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 41390 Mölndal, Sweden.,UK Dementia Research Institute at UCL, WC1E 6BT London, UK.,Department of Neurodegenerative Disease, UCL Institute of Neurology, WC1N 3BG London, UK
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, 08005 Barcelona, Spain.,Hospital del Mar Medical Research Institute (IMIM), 08005 Barcelona, Spain.,Universitat Pompeu Fabra, 08002 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBERBBN), 28089 Madrid, Spain
| | | |
Collapse
|
22
|
APOE in the normal brain. Neurobiol Dis 2020; 136:104724. [PMID: 31911114 DOI: 10.1016/j.nbd.2019.104724] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/19/2019] [Accepted: 12/31/2019] [Indexed: 12/25/2022] Open
Abstract
The APOE4 protein affects the primary neuropathological markers of Alzheimer's disease (AD): amyloid plaques, neurofibrillary tangles, and gliosis. These interactions have been investigated to understand the strong effect of APOE genotype on risk of AD. However, APOE genotype has strong effects on processes in normal brains, in the absence of the hallmarks of AD. We propose that CNS APOE is involved in processes in the normal brains that in later years apply specifically to processes of AD pathogenesis. We review the differences of the APOE protein found in the CNS compared to the plasma, including post-translational modifications (glycosylation, lipidation, multimer formation), focusing on ways that the common APOE isoforms differ from each other. We also review structural and functional studies of young human brains and control APOE knock-in mouse brains. These approaches demonstrate the effects of APOE genotype on microscopic neuron structure, gross brain structure, and behavior, primarily related to the hippocampal areas. By focusing on the effects of APOE genotype on normal brain function, approaches can be pursued to identify biomarkers of APOE dysfunction, to promote normal functions of the APOE4 isoform, and to prevent the accumulation of the pathologic hallmarks of AD with aging.
Collapse
|
23
|
Lamballais S, Muetzel RL, Ikram MA, Tiemeier H, Vernooij MW, White T, Adams HHH. Genetic Burden for Late-Life Neurodegenerative Disease and Its Association With Early-Life Lipids, Brain, Behavior, and Cognition. Front Psychiatry 2020; 11:33. [PMID: 32116848 PMCID: PMC7018686 DOI: 10.3389/fpsyt.2020.00033] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 01/10/2020] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Genetics play a significant role in the etiology of late-life neurodegenerative diseases like Alzheimer's disease, Parkinson's disease, and frontotemporal dementia. Part of the individual differences in risk for these diseases can be traced back decades before the onset of disease symptoms. Previous studies have shown evidence for plausible links of apolipoprotein E (APOE), the most important genetic marker for Alzheimer's disease, with early-life cognition and neuroimaging markers. We aimed to assess whether genome-wide genetic burden for the aforementioned neurodegenerative diseases plays a role in early-life processes. METHODS We studied children from the Generation R Study, a prospective birth cohort. APOE genotypes and polygenic genetic burdens for Alzheimer's disease, Parkinson's disease, and frontotemporal dementia were obtained through genome-wide genotyping. Non-verbal intelligence was assessed through cognitive tests at the research center around the age of 6 years, and educational attainment through a national school performance test around the age of 11 years. The Child Behavior Checklist was administered around the age of 10 years, and data from the anxious/depressed, withdrawn/depressed, and the internalizing behavior problems scales were used. Children participated in a neuroimaging study when they were 10 years old, in which structural brain metrics were obtained. Lipid serum profiles, which may be influenced by APOE genotype, were assessed from venal blood obtained around the age of 6 years. The sample size per analysis varied between 1,641 and 3,650 children due to completeness of data. RESULTS We did not find evidence that APOE genotype or the polygenic scores impact on childhood nonverbal intelligence, educational attainment, internalizing behavior, and global brain structural measures including total brain volume and whole brain fractional anisotropy (all p > 0.05). Carriership of the APOE ε2 allele was associated with lower and APOE ε4 with higher low-density lipoprotein cholesterol concentrations when compared to APOE ε3/ε3 carriers. CONCLUSION We found no evidence that genetic burden for late-life neurodegenerative diseases associates with early-life cognition, internalizing behavior, or global brain structure.
Collapse
Affiliation(s)
- Sander Lamballais
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Ryan L Muetzel
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands.,Department of Child and Adolescent Psychiatry and Psychology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Mohammad Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Henning Tiemeier
- Department of Child and Adolescent Psychiatry and Psychology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands.,Department of Social and Behavioral Science, Harvard T. H. Chan School of Public Health, Boston, MA, United States
| | - Meike W Vernooij
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands.,Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Tonya White
- Department of Child and Adolescent Psychiatry and Psychology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands.,Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Hieab H H Adams
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands.,Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands.,Department of Clinical Genetics, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
24
|
Miller ZA, Rosenberg L, Santos-Santos MA, Stephens M, Allen IE, Hubbard HI, Cantwell A, Mandelli ML, Grinberg LT, Seeley WW, Miller BL, Rabinovici GD, Gorno-Tempini ML. Prevalence of Mathematical and Visuospatial Learning Disabilities in Patients With Posterior Cortical Atrophy. JAMA Neurol 2019; 75:728-737. [PMID: 29630699 DOI: 10.1001/jamaneurol.2018.0395] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Importance Increased prevalence of language-based learning disabilities (LDs) has been previously reported in patients with primary progressive aphasia (PPA). This study hypothesized that patients with focal neurodegenerative syndromes outside the language network, such as posterior cortical atrophy (PCA), would have a higher rate of nonlanguage LDs, congruent with their mainly visuospatial presentation. Objective To investigate the prevalence and type of LD (language and/or mathematical and visuospatial) in a large cohort of patients with PCA compared with patients with logopenic variant PPA (lvPPA) and amnestic Alzheimer disease (AD). Design, Setting, and Participants This case-control study reviewed 279 medical records from a university-based clinic and research center for patients with neurodegenerative diseases for LD history, including patients with PCA (n = 95), patients with lvPPA (n = 84), and a matched cohort with amnestic AD (n = 100). No records were excluded. The study compared cognitive and neuroimaging features of patients with PCA with and without LDs. A review of the records of patients presenting from March 1, 1999, to August 31, 2014, revealed 95 PCA cases and 84 lvPPA cases. Then 100 patients with amnestic AD from this same period were chosen for comparison, matching against the groups for age, sex, and disease severity. Data analysis was performed from September 8, 2013, to November 6, 2017. Main Outcomes and Measures Prevalence of total LD history and prevalence of language and mathematical or visuospatial LD history across all cohorts. Results A total of 179 atypical AD cases (95 with PCA and 84 with lvPPA) and 100 disease control cases (amnestic AD) were included in the study. The groups were not statistically different for mean (SD) age at first visit (PCA, 61.9 [7.0] years; lvPPA, 65.1 [8.7] years; amnestic AD, 64.0 [12.6] years; P = .08), mean (SD) age at first symptom (PCA, 57.5 [7.0] years; lvPPA, 61.1 [9.0] years; amnestic AD, 59.6 [13.7] years; P = .06), or sex (PCA, 66.3% female; lvPPA, 56.0% female; amnestic AD, 57.0% female; P = .30) but differed on non-right-hand preference (PCA, 18.3%; lvPPA, 20.2%; amnestic AD, 7.7%; P = .04), race/ethnicity (PCA, 88.3% white; lvPPA, 99.0% white; amnestic AD, 80.0% white; P < .001), and mean (SD) educational level (PCA, 15.7 [3.2] years; lvPPA, 16.2 [3.3] years; amnestic AD, 14.8 [3.5] years; P = .02). A total of 18 of the 95 patients with PCA (18.9%) reported a history of LD, which is greater than the 3 of 100 patients (3.0%) in the amnestic AD cohort (P < .001) and the 10.0% expected rate in the general population (P = .007). In the PCA cohort, 13 of 95 patients (13.7%) had a nonlanguage mathematical and/or visuospatial LD; this rate was greater than that in the amnestic AD (1 of 100 [1.0%]; P < .001) and lvPPA (2 of 84 [2.4%]; P = .006) cohorts and greater than the 6.0% expected general population rate of mathematical LD (P = .003). Compared with the patients with PCA without LDs, the group with LDs had greater preservation of global cognition and a more right-lateralized pattern of atrophy. Conclusions and Relevance Nonlanguage mathematical and visuospatial LDs were associated with focal, visuospatial predominant neurodegenerative clinical syndromes. This finding supports the hypothesis that neurodevelopmental differences in specific brain networks are associated with phenotypic manifestation of later-life neurodegenerative disease.
Collapse
Affiliation(s)
- Zachary A Miller
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | - Lynne Rosenberg
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | - Miguel A Santos-Santos
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | - Melanie Stephens
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | - Isabel E Allen
- Department of Biostatistics, University of California, San Francisco
| | - H Isabel Hubbard
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | - Averill Cantwell
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | - Maria Luisa Mandelli
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | - Lea T Grinberg
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco.,Department of Pathology, University of California, San Francisco
| | - William W Seeley
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco.,Department of Pathology, University of California, San Francisco
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | - Gil D Rabinovici
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| | - Maria Luisa Gorno-Tempini
- Memory and Aging Center, Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco
| |
Collapse
|
25
|
Abstract
Technologies for imaging the pathophysiology of Alzheimer disease (AD) now permit studies of the relationships between the two major proteins deposited in this disease - amyloid-β (Aβ) and tau - and their effects on measures of neurodegeneration and cognition in humans. Deposition of Aβ in the medial parietal cortex appears to be the first stage in the development of AD, although tau aggregates in the medial temporal lobe (MTL) precede Aβ deposition in cognitively healthy older people. Whether aggregation of tau in the MTL is the first stage in AD or a fairly benign phenomenon that may be transformed and spread in the presence of Aβ is a major unresolved question. Despite a strong link between Aβ and tau, the relationship between Aβ and neurodegeneration is weak; rather, it is tau that is associated with brain atrophy and hypometabolism, which, in turn, are related to cognition. Although there is support for an interaction between Aβ and tau resulting in neurodegeneration that leads to dementia, the unknown nature of this interaction, the strikingly different patterns of brain Aβ and tau deposition and the appearance of neurodegeneration in the absence of Aβ and tau are challenges to this model that ultimately must be explained.
Collapse
|
26
|
Genetic predisposition, modifiable-risk-factor profile and long-term dementia risk in the general population. Nat Med 2019; 25:1364-1369. [PMID: 31451782 PMCID: PMC6739225 DOI: 10.1038/s41591-019-0547-7] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 07/12/2019] [Indexed: 01/26/2023]
Abstract
The exact etiology of dementia is still unclear, but both genetic and lifestyle factors are thought to be key drivers of this complex disease. The recognition of familial patterns of dementia has led to the discovery of genetic factors that have a role in the pathogenesis of dementia, including the apolipoprotein E (APOE) genotype and a large and still-growing number of genetic variants1,2. Beyond genetic architecture, several modifiable risk factors have been implicated in the development of dementia3. Prevention trials of measures to halt or delay cognitive decline are increasingly recruiting older individuals who are genetically predisposed to dementia. However, it remains unclear whether targeted health and lifestyle interventions can attenuate or even offset increased genetic risk. Here, we leverage long-term data on both genetic and modifiable risk factors from 6,352 individuals aged 55 years and older in the population-based Rotterdam Study. In this study, we demonstrate that, in individuals at low and intermediate genetic risk, favorable modifiable-risk profiles are related to a lower risk of dementia compared to unfavorable profiles. In contrast, these protective associations were not found in those at high genetic risk.
Collapse
|
27
|
Reynolds CA, Smolen A, Corley RP, Munoz E, Friedman NP, Rhee SH, Stallings MC, DeFries JC, Wadsworth SJ. APOE effects on cognition from childhood to adolescence. Neurobiol Aging 2019; 84:239.e1-239.e8. [PMID: 31126628 DOI: 10.1016/j.neurobiolaging.2019.04.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 04/13/2019] [Accepted: 04/14/2019] [Indexed: 01/10/2023]
Abstract
The ε4 allele of APOE is a well-established genetic risk factor for cognitive aging and dementia, but its influence on early life cognition is unknown. Consequently, we assessed associations of APOE genotypes with cognitive performance during 7, 12, and 16 year-assessments in our ongoing Colorado Adoption/Twin Study of Lifespan behavioral development (CATSLife). In general, APOE ε4 was associated with lower Verbal, Performance, and Full Scale IQ scores during childhood and adolescence (e.g., Full Scale IQ was lower by 1.91 points per ε4 allele, d = -0.13), with larger effects in females (e.g., average Full Scale IQ scores were 3.41 points lower in females per each ε4 allele vs. 0.33 points lower in males). Thus, these results suggest that deleterious effects of the APOE ε4 allele are manifested before adulthood, especially in females, and support both early origin theories and differential life-course vulnerabilities for later cognitive impairment.
Collapse
Affiliation(s)
- Chandra A Reynolds
- Department of Psychology, University of California, Riverside, Riverside, CA, USA.
| | - Andrew Smolen
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
| | - Robin P Corley
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
| | - Elizabeth Munoz
- Department of Psychology, University of California, Riverside, Riverside, CA, USA
| | - Naomi P Friedman
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
| | - Soo Hyun Rhee
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
| | - Michael C Stallings
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
| | - John C DeFries
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
| | - Sally J Wadsworth
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
| |
Collapse
|
28
|
APOE-ε4 risk variant for Alzheimer's disease modifies the association between cognitive performance and cerebral morphology in healthy middle-aged individuals. NEUROIMAGE-CLINICAL 2019; 23:101818. [PMID: 30991302 PMCID: PMC6463204 DOI: 10.1016/j.nicl.2019.101818] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 04/01/2019] [Accepted: 04/07/2019] [Indexed: 12/18/2022]
Abstract
The APOE-ε4 genotype is the highest genetic risk factor for Alzheimer's disease (AD). In cognitively unimpaired individuals, it has been related to altered brain morphology, function and earlier amyloid beta accumulation. However, its impact on cognitive performance is less evident. Here, we examine the impact of APOE-ε4 allele load in modulating the association between cognitive functioning and brain morphology in middle-aged healthy individuals. A high-resolution structural MRI scan was acquired and episodic memory (EM) as well as executive functions (EFs) were assessed in a sample of 527 middle-aged unimpaired individuals hosting a substantial representation of ε4-homozygous (N = 64). We adopted a voxel-wise unbiased method to assess whether the number of APOE-ε4 alleles significantly modified the associations between gray matter volumes (GMv) and performance in both cognitive domains. Even though the APOE-ε4 allele load did not exert a direct impact on any cognitive measures, it reversed the relationships between GMv and cognitive performance in a highly symmetrical topological pattern. For EM, interactions mapped onto the inferior temporal gyrus and the dorsal anterior cingulate cortex. Regarding EFs, significant interactions were observed for processing speed, working memory, and visuospatial attention in distinct brain regions. These results suggest that APOE-ε4 carriers display a structure-function association corresponding to an older age than their chronological one. Our findings additionally indicate that APOE-ε4 carriers may rely on the integrity of multiple compensatory brain systems in order to preserve their cognitive abilities, possibly due to an incipient neurodegeneration. Overall this study provides novel insights on the mechanisms through which APOE-ε4 posits an increased AD risk.
Collapse
|
29
|
Kalvas LB. The Life Course Health Development Model: A theoretical research framework for paediatric delirium. J Clin Nurs 2019; 28:2351-2360. [PMID: 30653772 DOI: 10.1111/jocn.14776] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 12/05/2018] [Accepted: 01/07/2019] [Indexed: 12/31/2022]
Abstract
AIMS AND OBJECTIVES To create a framework for future research through application and critique of the Life Course Health Development Model to the phenomenon of paediatric delirium. BACKGROUND Delirium in the paediatric intensive care unit is associated with increased duration of mechanical ventilation, length of stay and mortality. Nurses are uniquely positioned at the bedside to identify, prevent and treat delirium. An understanding of the potential long-term consequences of paediatric delirium is necessary to provide impetus for nursing research and practice change. The Life Course Health Development Model is a valuable tool when considering the multiple mechanisms and processes through which the experience of delirium could affect a child's life trajectory. DESIGN Critical review of the literature through application and critique of the Life Course Health Development Model in the context of paediatric delirium. Gaps in the current understanding of paediatric delirium, as well as future directions for research and practice, are discussed. METHODS The seven core principles of the model are considered in the context of paediatric delirium. Each of the principles has the potential to further understanding of paediatric delirium and identify areas for future inquiry. This discussion leads to a critique of the ability of the model to lead future research and practice change. CONCLUSIONS The Life Course Health Development Model depicts a process in which the acute and severe stress of critical illness leads to maladaptive neurologic changes that contribute to the development of delirium and impair a child's life trajectory. RELEVANCE TO CLINICAL PRACTICE By emphasising the potential lifelong consequences for critically ill children who experience delirium, this application of the Life Course Health Development Model will stimulate discussion, research and practice change among paediatric clinicians and researchers.
Collapse
|
30
|
Abstract
Population-based clinic-pathological studies have established that the most common pathological substrate of dementia in community-dwelling elderly people is mixed, especially Alzheimer's disease (AD) and cerebrovascular ischemic disease (CVID), rather than pure AD. While these could be just two frequent unrelated comorbidities in the elderly, epidemiological research has reinforced the idea that mid-life (age <65 years) vascular risk factors increase the risk of late-onset (age ≥ 65 years) dementia, and specifically AD. By contrast, healthy lifestyle choices such as leisure activities, physical exercise, and Mediterranean diet are considered protective against AD. Remarkably, several large population-based longitudinal epidemiological studies have recently indicated that the incidence and prevalence of dementia might be decreasing in Western countries. Although it remains unclear whether these positive trends are attributable to neuropathologically definite AD versus CVID, based on these epidemiological data it has been estimated that a sizable proportion of AD cases could be preventable. In this review, we discuss the current evidence about modifiable risk factors for AD derived from epidemiological, preclinical, and interventional studies, and analyze the opportunities for therapeutic and preventative interventions.
Collapse
Affiliation(s)
- Alberto Serrano-Pozo
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - John H. Growdon
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
31
|
Bussy A, Snider BJ, Coble D, Xiong C, Fagan AM, Cruchaga C, Benzinger TLS, Gordon BA, Hassenstab J, Bateman RJ, Morris JC. Effect of apolipoprotein E4 on clinical, neuroimaging, and biomarker measures in noncarrier participants in the Dominantly Inherited Alzheimer Network. Neurobiol Aging 2018; 75:42-50. [PMID: 30530186 DOI: 10.1016/j.neurobiolaging.2018.10.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 09/28/2018] [Accepted: 10/05/2018] [Indexed: 11/19/2022]
Abstract
The apolipoprotein E ε4 allele (APOE4) is the major genetic risk factor for sporadic Alzheimer's disease (AD). APOE4 may have effects on cognition and brain atrophy years before the onset of symptomatic AD. We analyzed the effects of APOE4 in a unique cohort of young adults who had undergone comprehensive assessments as part of the Dominantly Inherited Alzheimer Network (DIAN), an international longitudinal study of individuals from families with autosomal dominant AD. We analyzed the effect of an APOE4 allele on cognitive measures, volumetric MRI, amyloid deposition, glucose metabolism, and on cerebrospinal fluid levels of AD biomarkers in 162 participants that did not carry the mutant gene (noncarriers). APOE4+ and APOE4- mutation noncarriers had similar performance on cognitive measures. Amyloid deposition began at an earlier age in APOE4+ participants, whereas hippocampal volume was similar between the groups. These preliminary findings are consistent with growing evidence that the APOE4 allele may exert effects in midlife years before symptom onset, promoting amyloid deposition before altering cognitive performance or brain structure.
Collapse
Affiliation(s)
- Aurélie Bussy
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO; Department of Neurology, Washington University School of Medicine, Saint Louis, MO
| | - B Joy Snider
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO; Department of Neurology, Washington University School of Medicine, Saint Louis, MO.
| | - Dean Coble
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO; Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO
| | - Chengjie Xiong
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO; Division of Biostatistics, Washington University School of Medicine, Saint Louis, MO
| | - Anne M Fagan
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO; Department of Neurology, Washington University School of Medicine, Saint Louis, MO
| | - Carlos Cruchaga
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO; Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO
| | - Tammie L S Benzinger
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO; Department of Radiology, Washington University School of Medicine, Saint Louis, MO
| | - Brian A Gordon
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO; Department of Radiology, Washington University School of Medicine, Saint Louis, MO
| | - Jason Hassenstab
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO; Department of Neurology, Washington University School of Medicine, Saint Louis, MO
| | - Randall J Bateman
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO; Department of Neurology, Washington University School of Medicine, Saint Louis, MO
| | - John C Morris
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, Saint Louis, MO; Department of Neurology, Washington University School of Medicine, Saint Louis, MO
| |
Collapse
|
32
|
Viswanath B, Rao NP, Narayanaswamy JC, Sivakumar PT, Kandasamy A, Kesavan M, Mehta UM, Venkatasubramanian G, John JP, Mukherjee O, Purushottam M, Kannan R, Mehta B, Kandavel T, Binukumar B, Saini J, Jayarajan D, Shyamsundar A, Moirangthem S, Vijay Kumar KG, Thirthalli J, Chandra PS, Gangadhar BN, Murthy P, Panicker MM, Bhalla US, Chattarji S, Benegal V, Varghese M, Reddy JYC, Raghu P, Rao M, Jain S. Discovery biology of neuropsychiatric syndromes (DBNS): a center for integrating clinical medicine and basic science. BMC Psychiatry 2018; 18:106. [PMID: 29669557 PMCID: PMC5907468 DOI: 10.1186/s12888-018-1674-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 03/21/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND There is emerging evidence that there are shared genetic, environmental and developmental risk factors in psychiatry, that cut across traditional diagnostic boundaries. With this background, the Discovery biology of neuropsychiatric syndromes (DBNS) proposes to recruit patients from five different syndromes (schizophrenia, bipolar disorder, obsessive compulsive disorder, Alzheimer's dementia and substance use disorders), identify those with multiple affected relatives, and invite these families to participate in this study. The families will be assessed: 1) To compare neuro-endophenotype measures between patients, first degree relatives (FDR) and healthy controls., 2) To identify cellular phenotypes which differentiate the groups., 3) To examine the longitudinal course of neuro-endophenotype measures., 4) To identify measures which correlate with outcome, and 5) To create a unified digital database and biorepository. METHODS The identification of the index participants will occur at well-established specialty clinics. The selected individuals will have a strong family history (with at least another affected FDR) of mental illness. We will also recruit healthy controls without family history of such illness. All recruited individuals (N = 4500) will undergo brief clinical assessments and a blood sample will be drawn for isolation of DNA and peripheral blood mononuclear cells (PBMCs). From among this set, a subset of 1500 individuals (300 families and 300 controls) will be assessed on several additional assessments [detailed clinical assessments, endophenotype measures (neuroimaging- structural and functional, neuropsychology, psychophysics-electroencephalography, functional near infrared spectroscopy, eye movement tracking)], with the intention of conducting repeated measurements every alternate year. PBMCs from this set will be used to generate lymphoblastoid cell lines, and a subset of these would be converted to induced pluripotent stem cell lines and also undergo whole exome sequencing. DISCUSSION We hope to identify unique and overlapping brain endophenotypes for major psychiatric syndromes. In a proportion of subjects, we expect these neuro-endophenotypes to progress over time and to predict treatment outcome. Similarly, cellular assays could differentiate cell lines derived from such groups. The repository of biomaterials as well as digital datasets of clinical parameters, will serve as a valuable resource for the broader scientific community who wish to address research questions in the area.
Collapse
Affiliation(s)
- Biju Viswanath
- National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | - Naren P. Rao
- National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | | | | | - Arun Kandasamy
- National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | - Muralidharan Kesavan
- National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | | | | | - John P. John
- National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | - Odity Mukherjee
- Institute for Stem Cell Biology and Regenerative Medicine (InStem), Bangalore, India
| | - Meera Purushottam
- National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | - Ramakrishnan Kannan
- National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | - Bhupesh Mehta
- National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | - Thennarasu Kandavel
- National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | - B. Binukumar
- National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | - Jitender Saini
- National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | - Deepak Jayarajan
- National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | - A. Shyamsundar
- National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | - Sydney Moirangthem
- National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | - K. G. Vijay Kumar
- National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | - Jagadisha Thirthalli
- National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | - Prabha S. Chandra
- National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | | | - Pratima Murthy
- National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | - Mitradas M. Panicker
- National Centre for Biological Sciences, Tata Institute of Fundamental Research (NCBS-TIFR), Bangalore, India
| | - Upinder S. Bhalla
- National Centre for Biological Sciences, Tata Institute of Fundamental Research (NCBS-TIFR), Bangalore, India
| | - Sumantra Chattarji
- Institute for Stem Cell Biology and Regenerative Medicine (InStem), Bangalore, India
- National Centre for Biological Sciences, Tata Institute of Fundamental Research (NCBS-TIFR), Bangalore, India
| | - Vivek Benegal
- National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | - Mathew Varghese
- National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | | | - Padinjat Raghu
- National Centre for Biological Sciences, Tata Institute of Fundamental Research (NCBS-TIFR), Bangalore, India
| | - Mahendra Rao
- Institute for Stem Cell Biology and Regenerative Medicine (InStem), Bangalore, India
| | - Sanjeev Jain
- National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| |
Collapse
|
33
|
Myrum C, Nikolaienko O, Bramham CR, Haavik J, Zayats T. Implication of the APP Gene in Intellectual Abilities. J Alzheimers Dis 2018; 59:723-735. [PMID: 28671113 PMCID: PMC5523840 DOI: 10.3233/jad-170049] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background: Cognitive functions are highly heritable and polygenic, though the source of this genetic influence is unclear. On the neurobiological level, these functions rely on effective neuroplasticity, in which the activity-regulated cytoskeleton associated protein (ARC) plays an essential role. Objectives: To examine whether the ARC gene complex may contribute to the genetic components of intellectual function given the crucial role of ARC in brain plasticity and memory formation. Methods: The ARC complex was tested for association with intelligence (IQ) in children from the Avon Longitudinal Study of Parents and Children (ALSPAC, N = 5,165). As Alzheimer’s disease (AD) shares genetics with cognitive functioning, the association was followed up in an AD sample (17,008 cases, 37,154 controls). Results: The ARC complex revealed association with verbal and total IQ (empirical p = 0.027 and 0.041, respectively) in the ALSPAC. The strongest single variant signal (rs2830077; empirical p = 0.018), within the APP gene, was confirmed in the AD sample (p = 2.76E-03). Functional analyses of this variant showed its preferential binding to the transcription factor CP2. Discussion: This study implicates APP in childhood IQ. While follow-up studies are needed, this observation could help elucidate the etiology of disorders associated with cognitive dysfunction, such as AD.
Collapse
Affiliation(s)
- Craig Myrum
- K.G. Jebsen Centre for Neuropsychiatric Disorders, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Oleksii Nikolaienko
- K.G. Jebsen Centre for Neuropsychiatric Disorders, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Clive R Bramham
- K.G. Jebsen Centre for Neuropsychiatric Disorders, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Jan Haavik
- K.G. Jebsen Centre for Neuropsychiatric Disorders, Department of Biomedicine, University of Bergen, Bergen, Norway.,Department of Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Tetyana Zayats
- K.G. Jebsen Centre for Neuropsychiatric Disorders, Department of Biomedicine, University of Bergen, Bergen, Norway
| |
Collapse
|
34
|
Oishi K, Chang L, Huang H. Baby brain atlases. Neuroimage 2018; 185:865-880. [PMID: 29625234 DOI: 10.1016/j.neuroimage.2018.04.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 02/27/2018] [Accepted: 04/02/2018] [Indexed: 01/23/2023] Open
Abstract
The baby brain is constantly changing due to its active neurodevelopment, and research into the baby brain is one of the frontiers in neuroscience. To help guide neuroscientists and clinicians in their investigation of this frontier, maps of the baby brain, which contain a priori knowledge about neurodevelopment and anatomy, are essential. "Brain atlas" in this review refers to a 3D-brain image with a set of reference labels, such as a parcellation map, as the anatomical reference that guides the mapping of the brain. Recent advancements in scanners, sequences, and motion control methodologies enable the creation of various types of high-resolution baby brain atlases. What is becoming clear is that one atlas is not sufficient to characterize the existing knowledge about the anatomical variations, disease-related anatomical alterations, and the variations in time-dependent changes. In this review, the types and roles of the human baby brain MRI atlases that are currently available are described and discussed, and future directions in the field of developmental neuroscience and its clinical applications are proposed. The potential use of disease-based atlases to characterize clinically relevant information, such as clinical labels, in addition to conventional anatomical labels, is also discussed.
Collapse
Affiliation(s)
- Kenichi Oishi
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Linda Chang
- Departments of Diagnostic Radiology and Nuclear Medicine, and Neurology, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Medicine, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Hao Huang
- Department of Radiology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA; Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
35
|
Madan CR. Advances in Studying Brain Morphology: The Benefits of Open-Access Data. Front Hum Neurosci 2017; 11:405. [PMID: 28824407 PMCID: PMC5543094 DOI: 10.3389/fnhum.2017.00405] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 07/21/2017] [Indexed: 12/20/2022] Open
|
36
|
Nao J, Sun H, Wang Q, Ma S, Zhang S, Dong X, Ma Y, Wang X, Zheng D. Adverse Effects of the Apolipoprotein E ε4 Allele on Episodic Memory, Task Switching and Gray Matter Volume in Healthy Young Adults. Front Hum Neurosci 2017; 11:346. [PMID: 28706481 PMCID: PMC5489690 DOI: 10.3389/fnhum.2017.00346] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 06/15/2017] [Indexed: 11/13/2022] Open
Abstract
Many studies have shown that healthy elderly subjects and patients with Alzheimer's disease (AD) who carry the apolipoprotein E (ApoE) ε4 allele have worse cognitive function and more severe brain atrophy than non-carriers. However, it remains unclear whether this ApoE polymorphism leads to changes of cognition and brain morphology in healthy young adults. In this study, we used an established model to measure verbal episodic memory and core executive function (EF) components (response inhibition, working memory and task switching) in 32 ApoE ε4 carriers and 40 non-carriers between 20 years and 40 years of age. To do this, we carried out an adapted auditory verbal learning test and three computerized EF tasks. High-resolution head magnetic resonance scans were performed in all participants and voxel-based morphometry (VBM) was used for image processing and analysis. Multivariate analysis of variance (ANOVA) performed on memory measures showed that the overall verbal episodic memory of ApoE ε4 carriers was significantly worse than non-carriers (Wilk's λ = 4.884, P = 0.004). No significant differences were detected in overall EF between the two groups. Post hoc analyses revealed group differences in terms of immediate recall, recognition and task switching, which favored non-carriers. VBM analysis showed gray matter (GM) bilateral reductions in the medial and dorsolateral frontal, parietal and left temporal cortices in the carrier group relative to the non-carrier group, which were most significant in the bilateral anterior and middle cingulate gyri. However, these changes in GM volume were not directly associated with changes in cognitive function. Our data show that the ApoE ε4 allele is associated with poorer performance in verbal episodic memory and task switching, and a reduction in GM volume in healthy young adults, suggesting that the effects of ApoE ε4 upon cognition and brain morphology exist long before the possible occurrence of AD.
Collapse
Affiliation(s)
- Jianfei Nao
- Department of Neurology, Shengjing Hospital of China Medical UniversityShenyang, China
| | - Hongzan Sun
- Department of Radiology, Shengjing Hospital of China Medical UniversityShenyang, China
| | - Qiushi Wang
- Department of Radiology, Shengjing Hospital of China Medical UniversityShenyang, China
| | - Shuang Ma
- Department of Neurology, Shengjing Hospital of China Medical UniversityShenyang, China
| | - Shuo Zhang
- Department of Neurology, Shengjing Hospital of China Medical UniversityShenyang, China
| | - Xiaoyu Dong
- Department of Neurology, Shengjing Hospital of China Medical UniversityShenyang, China
| | - Ying Ma
- Department of Neurology, Shengjing Hospital of China Medical UniversityShenyang, China
| | - Xiaoming Wang
- Department of Radiology, Shengjing Hospital of China Medical UniversityShenyang, China
| | - Dongming Zheng
- Department of Neurology, Shengjing Hospital of China Medical UniversityShenyang, China
| |
Collapse
|
37
|
Huntington Disease as a Neurodevelopmental Disorder and Early Signs of the Disease in Stem Cells. Mol Neurobiol 2017; 55:3351-3371. [PMID: 28497201 PMCID: PMC5842500 DOI: 10.1007/s12035-017-0477-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/01/2017] [Indexed: 02/07/2023]
Abstract
Huntington disease (HD) is a dominantly inherited disorder caused by a CAG expansion mutation in the huntingtin (HTT) gene, which results in the HTT protein that contains an expanded polyglutamine tract. The adult form of HD exhibits a late onset of the fully symptomatic phase. However, there is also a long presymptomatic phase, which has been increasingly investigated and recognized as important for the disease development. Moreover, the juvenile form of HD, evoked by a higher number of CAG repeats, resembles a neurodevelopmental disorder and has recently been the focus of additional interest. Multiple lines of data, such as the developmental necessity of HTT, its role in the cell cycle and neurogenesis, and findings from pluripotent stem cells, suggest the existence of a neurodevelopmental component in HD pathogenesis. Therefore, we discuss the early molecular pathogenesis of HD in pluripotent and neural stem cells, with respect to the neurodevelopmental aspects of HD.
Collapse
|
38
|
Developmental alterations in Huntington's disease neural cells and pharmacological rescue in cells and mice. Nat Neurosci 2017; 20:648-660. [PMID: 28319609 PMCID: PMC5610046 DOI: 10.1038/nn.4532] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 02/23/2017] [Indexed: 12/11/2022]
Abstract
Neural cultures derived from Huntington's disease (HD) patient-derived induced pluripotent stem cells were used for 'omics' analyses to identify mechanisms underlying neurodegeneration. RNA-seq analysis identified genes in glutamate and GABA signaling, axonal guidance and calcium influx whose expression was decreased in HD cultures. One-third of gene changes were in pathways regulating neuronal development and maturation. When mapped to stages of mouse striatal development, the profiles aligned with earlier embryonic stages of neuronal differentiation. We observed a strong correlation between HD-related histone marks, gene expression and unique peak profiles associated with dysregulated genes, suggesting a coordinated epigenetic program. Treatment with isoxazole-9, which targets key dysregulated pathways, led to amelioration of expanded polyglutamine repeat-associated phenotypes in neural cells and of cognitive impairment and synaptic pathology in HD model R6/2 mice. These data suggest that mutant huntingtin impairs neurodevelopmental pathways that could disrupt synaptic homeostasis and increase vulnerability to the pathologic consequence of expanded polyglutamine repeats over time.
Collapse
|
39
|
Genetics: Alzheimer risk variants affect children's brains. Nat Rev Neurol 2016; 12:494. [PMID: 27469465 DOI: 10.1038/nrneurol.2016.115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|