1
|
Daly J, De Luca F, Berens SC, Field AP, Rusted JM, Bird CM. The effect of apolipoprotein E genotype on spatial processing in humans: A meta-analysis and systematic review. Cortex 2024; 177:268-284. [PMID: 38878339 DOI: 10.1016/j.cortex.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/04/2024] [Accepted: 05/20/2024] [Indexed: 07/31/2024]
Abstract
The ε4 allele of the apolipoprotein E (APOE4) gene is an established risk factor for Alzheimer's disease but its impact on cognition in healthy adults across the lifespan is unclear. One cognitive domain that is affected early in the course of Alzheimer's disease is spatial cognition, yet the evidence for APOE-related changes in spatial cognition is mixed. In this meta-analysis we assessed the impact of carrying the APOE4 allele on five subdomains of spatial cognition across the lifespan. We included studies of healthy human participants where an APOE4-carrier group (heterozygous or homozygous) could be compared to a homozygous group of APOE3-carriers. We identified 156 studies in total from three databases (Pubmed, Scopus and Web of Science) as well as through searching cited literature and contacting authors for unpublished data. 122 studies involving 32,547 participants were included in a meta-analysis, and the remaining studies are included in a descriptive review. APOE4 carriers scored significantly lower than APOE3 carriers (θˆ = -.08 [-.14, -.02]) on tests of spatial long-term memory; this effect was very small and was not modulated by age. On other subdomains of spatial cognition (spatial construction, spatial working memory, spatial reasoning, navigation) there were no effects of genotype. Overall, our results demonstrate that the APOE4 allele exerts little influence on spatial cognitive abilities in healthy adults.
Collapse
Affiliation(s)
- Jessica Daly
- Sussex Neuroscience, School of Psychology, University of Sussex, United Kingdom
| | - Flavia De Luca
- Sussex Neuroscience, School of Psychology, University of Sussex, United Kingdom
| | - Sam C Berens
- Sussex Neuroscience, School of Psychology, University of Sussex, United Kingdom
| | - Andy P Field
- School of Psychology, University of Sussex, United Kingdom
| | | | - Chris M Bird
- Sussex Neuroscience, School of Psychology, University of Sussex, United Kingdom.
| |
Collapse
|
2
|
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 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
|
3
|
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
|
4
|
Bousiges O, Cretin B, Muller C, Botzung A, Sanna L, Anthony P, Philippi N, Demuynck C, Blanc F. Involvement of ApoE4 in dementia with Lewy bodies in the prodromal and demented stages: evaluation of the Strasbourg cohort. GeroScience 2024; 46:1527-1542. [PMID: 37653269 PMCID: PMC10828291 DOI: 10.1007/s11357-023-00883-6] [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: 06/08/2023] [Accepted: 07/19/2023] [Indexed: 09/02/2023] Open
Abstract
ApoE4 as a risk factor for dementia with Lewy bodies (DLB) is still an issue. We sought to determine the involvement of ApoE4 according to different clinical parameters in our cohort of patients from Strasbourg, France. ApoE genotyping was performed on the AlphaLewyMA cohort. In this cohort, 197 patients were genotyped: 105 DLB patients, 37 Alzheimer's disease (AD) patients, 29 patients with AD/DLB comorbidity, and 26 control subjects (CS). The groups of patients were also classified according to the stage of evolution of the disease: prodromal or demented. We analyzed other parameters in relation to ApoE4 status, such as years of education (YOE) and Alzheimer CSF biomarkers. We observed a higher proportion of ApoE4 carriers in the AD (51.4%) and AD/DLB (72.4%) groups compared to the DLB (25.7%) and CS (11.5%) groups (p < 0.0001). We found a correlation between age at disease onset and YOE in the AD group (p = 0.039) but not in the DLB group (p = 0.056). Interestingly, in the DLB group, the subgroup of patients with high YOE (≥ 11) had significantly more patients with ApoE4 than the subgroup with low YOE (< 11). AD biomarkers did not seem to be impacted by the presence of ApoE4, except for Aβ42: DLB ApoE4-positive demented patients showed a more marked Aβ42 decrease. ApoE4 does not appear to be a risk factor for "pure" DLB patients. These results suggest a strong link between ApoE4 and amyloidopathy and consequently with AD. Trial registration: AlphaLewyMa, Identifier: NCT01876459, date of registration: June 12, 2013.
Collapse
Affiliation(s)
- Olivier Bousiges
- ICube Laboratory UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), IMIS Team and IMAGeS Team, University of Strasbourg and CNRS, Strasbourg, France.
- Laboratory of Biochemistry and Molecular Biology, University Hospital of Strasbourg, Strasbourg, France.
| | - Benjamin Cretin
- ICube Laboratory UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), IMIS Team and IMAGeS Team, University of Strasbourg and CNRS, Strasbourg, France
- CM2R (Research and Resources Memory Center), Geriatrics Department, Geriatric Day Hospital, Neurogeriatric Service, University Hospital of Strasbourg, Strasbourg, France
| | - Candice Muller
- CM2R (Research and Resources Memory Center), Geriatrics Department, Geriatric Day Hospital, Neurogeriatric Service, University Hospital of Strasbourg, Strasbourg, France
| | - Anne Botzung
- ICube Laboratory UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), IMIS Team and IMAGeS Team, University of Strasbourg and CNRS, Strasbourg, France
- CM2R (Research and Resources Memory Center), Geriatrics Department, Geriatric Day Hospital, Neurogeriatric Service, University Hospital of Strasbourg, Strasbourg, France
| | - Lea Sanna
- CM2R (Research and Resources Memory Center), Geriatrics Department, Geriatric Day Hospital, Neurogeriatric Service, University Hospital of Strasbourg, Strasbourg, France
| | - Pierre Anthony
- CM2R, Neuropsychology Unit, Head and Neck Department, Neurology Department, University of Strasbourg, Strasbourg, France
- CM2R, Geriatrics Department and Neurology Department, Colmar General Hospital, Colmar, France
| | - Nathalie Philippi
- ICube Laboratory UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), IMIS Team and IMAGeS Team, University of Strasbourg and CNRS, Strasbourg, France
- CM2R, Neuropsychology Unit, Head and Neck Department, Neurology Department, University of Strasbourg, Strasbourg, France
| | - Catherine Demuynck
- CM2R (Research and Resources Memory Center), Geriatrics Department, Geriatric Day Hospital, Neurogeriatric Service, University Hospital of Strasbourg, Strasbourg, France
| | - Frédéric Blanc
- ICube Laboratory UMR 7357 and FMTS (Fédération de Médecine Translationnelle de Strasbourg), IMIS Team and IMAGeS Team, University of Strasbourg and CNRS, Strasbourg, France
- CM2R (Research and Resources Memory Center), Geriatrics Department, Geriatric Day Hospital, Neurogeriatric Service, University Hospital of Strasbourg, Strasbourg, France
| |
Collapse
|
5
|
Topriceanu CC, Shah M, Webber M, Chan F, Shiwani H, Richards M, Schott J, Chaturvedi N, Moon JC, Hughes AD, Hingorani AD, O'Regan DP, Captur G. APOE ε4 carriage associates with improved myocardial performance from adolescence to older age. BMC Cardiovasc Disord 2024; 24:172. [PMID: 38509472 PMCID: PMC10956279 DOI: 10.1186/s12872-024-03808-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 02/21/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND Although APOE ε4 allele carriage confers a risk for coronary artery disease, its persistence in humans might be explained by certain survival advantages (antagonistic pleiotropy). METHODS Combining data from ~ 37,000 persons from three older age British cohorts (1946 National Survey of Health and Development [NSHD], Southall and Brent Revised [SABRE], and UK Biobank) and one younger age cohort (Avon Longitudinal Study of Parents and Children [ALSPAC]), we explored whether APOE ε4 carriage associates with beneficial or unfavorable left ventricular (LV) structural and functional metrics by echocardiography and cardiovascular magnetic resonance (CMR). RESULTS Compared to the non-APOE ε4 group, APOE ε4 carriers had similar cardiac phenotypes in terms of LV ejection fraction, E/e', posterior wall and interventricular septal thickness, and LV mass. However, they had improved myocardial performance resulting in greater LV stroke volume generation per 1 mL of myocardium (higher myocardial contraction fraction). In NSHD (n = 1467) and SABRE (n = 1187), ε4 carriers had a 4% higher MCF (95% CI 1-7%, p = 0.016) using echocardiography. Using CMR data, in UK Biobank (n = 32,972), ε4 carriers had a 1% higher MCF 95% (CI 0-1%, p = 0.020) with a dose-response relationship based on the number of ε4 alleles. In addition, UK Biobank ε4 carriers also had more favorable radial and longitudinal strain rates compared to non APOE ε4 carriers. In ALSPAC (n = 1397), APOE ε4 carriers aged < 24 years had a 2% higher MCF (95% CI 0-5%, p = 0.059). CONCLUSIONS By triangulating results in four independent cohorts, across imaging modalities (echocardiography and CMR), and in ~ 37,000 individuals, our results point towards an association between ε4 carriage and improved cardiac performance in terms of LV MCF. This potentially favorable cardiac phenotype adds to the growing number of reported survival advantages attributed to the pleiotropic effects APOE ε4 carriage that might collectively explain its persistence in human populations.
Collapse
Affiliation(s)
- Constantin-Cristian Topriceanu
- UCL MRC Unit for Lifelong Health and Ageing, University College London, London, UK
- UCL Institute of Cardiovascular Science, University College London, London, UK
- Cardiac MRI Unit, Barts Heart Centre, London, UK
- Cardiology Department, Centre for Inherited Heart Muscle Conditions, The Royal Free Hospital, Pond Street, Hampstead, London, UK
| | - Mit Shah
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London, UK
- MRC London Institute of Medical Science, Imperial College London, London, UK
| | - Matthew Webber
- UCL MRC Unit for Lifelong Health and Ageing, University College London, London, UK
- UCL Institute of Cardiovascular Science, University College London, London, UK
| | - Fiona Chan
- UCL MRC Unit for Lifelong Health and Ageing, University College London, London, UK
- UCL Institute of Cardiovascular Science, University College London, London, UK
| | - Hunain Shiwani
- UCL Institute of Cardiovascular Science, University College London, London, UK
- Cardiac MRI Unit, Barts Heart Centre, London, UK
| | - Marcus Richards
- UCL MRC Unit for Lifelong Health and Ageing, University College London, London, UK
| | - Jonathan Schott
- UCL MRC Unit for Lifelong Health and Ageing, University College London, London, UK
- Dementia Research Centre, UCL Queen Square Institute of Neurology, London, UK
| | - Nishi Chaturvedi
- UCL MRC Unit for Lifelong Health and Ageing, University College London, London, UK
- UCL Institute of Cardiovascular Science, University College London, London, UK
| | - James C Moon
- UCL Institute of Cardiovascular Science, University College London, London, UK
- Cardiac MRI Unit, Barts Heart Centre, London, UK
| | - Alun D Hughes
- UCL MRC Unit for Lifelong Health and Ageing, University College London, London, UK
- UCL Institute of Cardiovascular Science, University College London, London, UK
| | - Aroon D Hingorani
- UCL Institute of Cardiovascular Science, University College London, London, UK
- BHF Research Accelerator, University College London, London, UK
- Health Data Research, University College London, London, UK
| | - Declan P O'Regan
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London, UK
- MRC London Institute of Medical Science, Imperial College London, London, UK
| | - Gabriella Captur
- UCL MRC Unit for Lifelong Health and Ageing, University College London, London, UK.
- UCL Institute of Cardiovascular Science, University College London, London, UK.
- Cardiac MRI Unit, Barts Heart Centre, London, UK.
- Imperial Centre for Translational and Experimental Medicine, National Heart and Lung Institute, Imperial College London, London, UK.
- Cardiology Department, Centre for Inherited Heart Muscle Conditions, The Royal Free Hospital, Pond Street, Hampstead, London, UK.
| |
Collapse
|
6
|
Homayouni R, Daugherty AM, Yu Q, Raz N, Ofen N. KIBRA single nucleotide polymorphism is associated with hippocampal subfield volumes and cognition across development. Brain Struct Funct 2024; 229:223-230. [PMID: 37853296 DOI: 10.1007/s00429-023-02716-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 09/21/2023] [Indexed: 10/20/2023]
Abstract
The hippocampus (Hc) consists of cytoarchitectonically and functionally distinct subfields: dentate gyrus (DG), cornu ammonis (CA1-3), and subiculum. In adults, a single nucleotide polymorphism (rs17070145, C→ T) in KIBRA, a gene encoding the eponymous (KIdney-BRAin) protein, is associated with variability in Hc subfield volumes and episodic memory. T-allele carriers have larger DG and CA volumes and better episodic memory compared to C-homozygotes. Little is known, however, about KIBRA's role in the development of the brain and cognition. In a sample of children, adolescents, and young adults (N = 176, ages 5- 25 years), we replicated the adult association between KIBRA T-allele and larger DG and CA volumes but observed no relationship between KIBRA rs17070145 polymorphism and episodic memory. We noted, however, that a general cognitive performance index (IQ) differed across the allelic groups, with the lowest scores among T-homozygotes and the highest among C-homozygotes. Thus, in this developmental sample, KIBRA appears to have opposing effects on regional brain volume and cognition. These influences of KIBRA SNP may stem from associations between developmental reduction in brain volume and gains in cognitive performance-a hypothesis to be tested in longitudinal studies.
Collapse
Affiliation(s)
- Roya Homayouni
- Institute of Gerontology, Wayne State University, Detroit, MI, USA
- Department of Psychology, Wayne State University, Detroit, MI, USA
| | - Ana M Daugherty
- Institute of Gerontology, Wayne State University, Detroit, MI, USA
- Department of Psychology, Wayne State University, Detroit, MI, USA
| | - Qijing Yu
- Institute of Gerontology, Wayne State University, Detroit, MI, USA
| | - Naftali Raz
- Department of Psychology, Stony Brook University, Stony Brook, NY, USA
| | - Noa Ofen
- Institute of Gerontology, Wayne State University, Detroit, MI, USA.
- Department of Psychology, Wayne State University, Detroit, MI, USA.
- Merrill Palmer Skillman Institute, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
7
|
Scheinman SB, Tseng KY, Alford S, Tai LM. Higher Neuronal Facilitation and Potentiation with APOE4 Suppressed by Angiotensin II. Mol Neurobiol 2024; 61:120-131. [PMID: 37589833 PMCID: PMC10843153 DOI: 10.1007/s12035-023-03556-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/01/2023] [Indexed: 08/18/2023]
Abstract
Progressive hippocampal degeneration is a key component of Alzheimer's disease (AD) progression. Therefore, identifying how hippocampal neuronal function is modulated early in AD is an important approach to eventually prevent degeneration. AD-risk factors and signaling molecules likely modulate neuronal function, including APOE genotype and angiotensin II. Compared to APOE3, APOE4 increases AD risk up to 12-fold, and high levels of angiotensin II are hypothesized to disrupt neuronal function in AD. However, the extent that APOE and angiotensin II modulates the hippocampal neuronal phenotype in AD-relevant models is unknown. To address this issue, we used electrophysiological techniques to assess the impact of APOE genotype and angiotensin II on basal synaptic transmission, presynaptic, and post-synaptic activity in mice that express human APOE3 (E3FAD) or APOE4 (E4FAD) and overproduce Aβ. We found that compared to E3FAD mice, E4FAD mice have lower synaptic activity, but higher levels of paired-pulse facilitation (PPF) and long-term potentiation (LTP) in the Schaffer Collateral Commissural Pathway (SCCP) of the hippocampus. We also found that exogenous angiotensin II has a profound inhibitory effect on hippocampal LTP in both E3FAD and E4FAD mice. Collectively, our data suggests that APOE4 and Aβ are associated with a hippocampal phenotype comprised of lower basal activity and higher responses to high-frequency stimulation, the latter of which is suppressed by angiotensin II. These novel data suggest a potential mechanistic link between hippocampal activity, APOE4 genotype, and angiotensin II in AD.
Collapse
Affiliation(s)
- Sarah B Scheinman
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 808 S. Wood St. Rm 578 MC 512, Chicago, IL, 60612, USA
| | - Kuei Y Tseng
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 808 S. Wood St. Rm 578 MC 512, Chicago, IL, 60612, USA
| | - Simon Alford
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 808 S. Wood St. Rm 578 MC 512, Chicago, IL, 60612, USA
| | - Leon M Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, 808 S. Wood St. Rm 578 MC 512, Chicago, IL, 60612, USA.
| |
Collapse
|
8
|
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
|
9
|
Kellogg CM, Pham K, Ko S, Cox JEJ, Machalinski AH, Stout MB, Sharpe AL, Beckstead MJ, Chucair-Elliott AJ, Ocañas SR, Freeman WM. Consistent specificity and efficiency of tamoxifen-mediated cre induction across ages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.19.558482. [PMID: 37781585 PMCID: PMC10541132 DOI: 10.1101/2023.09.19.558482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Temporally controlling cre recombination through tamoxifen (Tam) induction has many advantages for biomedical research. Most studies report Tam induction at early post-natal/juvenile (<2 m.o.) mouse ages, but age-related neurodegeneration and aging studies can require cre induction in older mice (>12 m.o.). While anecdotally reported as problematic, there are no published comparisons of Tam mediated cre induction at early and late ages. Here, microglial-specific Cx3cr1 creERT 2 mice were crossed to a floxed NuTRAP reporter to compare cre induction at early (3-6 m.o.) and late (20 m.o.) ages. Specificity and efficiency of microglial labeling at 21-22 m.o. were identical in mice induced with Tam at 3-6 m.o. or 20 m.o. of age. Age-related microglial translatomic changes were also similar regardless of Tam induction age. Each cre and flox mouse line should be validated independently, however, these findings demonstrate that Tam-mediated cre induction can be performed even into older mouse ages.
Collapse
|
10
|
Pirraglia E, Glodzik L, Shao Y. Lower mortality risk in APOE4 carriers with normal cognitive ageing. Sci Rep 2023; 13:15089. [PMID: 37699966 PMCID: PMC10497512 DOI: 10.1038/s41598-023-41078-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/21/2023] [Indexed: 09/14/2023] Open
Abstract
Abnormal cognitive ageing, including dementia, poses serious challenges to health and social systems in ageing populations. As such, characterizing factors associated with abnormal cognitive ageing and developing needed preventive measures are of great importance. The ε4 allele of the Apolipoprotein E gene (APOE4) is a well-known genetic risk factor for late-onset Alzheimer's disease. APOE4 carriers are also at elevated risk of cardiovascular diseases which are associated with increased risk of cognitive impairment. On the other hand, APOE4 is known to be associated with reduced risk of multiple common types of cancer-a major age-related disease and leading cause of mortality. We conducted the first-ever study of APOE4's opposing effects on cognitive decline and mortality using competing risk models considering two types of death-death with high-amounts versus low-amounts of autopsy-assessed Alzheimer's neuropathology. We observed that APOE4 was associated with decreased mortality risk in people who died with low amounts of Alzheimer's-type neuropathology, but APOE4 was associated with increased mortality risk in people who died with high amounts of Alzheimer's-type neuropathology, a major risk factor of cognitive impairment. Possible preventive measures of abnormal cognitive ageing are also discussed.
Collapse
Affiliation(s)
- Elizabeth Pirraglia
- Department of Population Health, New York University Grossman School of Medicine, New York, NY, USA
| | - Lidia Glodzik
- Department of Radiology, Weill Cornell Medicine, Brain Health Imaging Institute, New York, NY, USA
| | - Yongzhao Shao
- Department of Population Health, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
11
|
Langella S, Barksdale NG, Vasquez D, Aguillon D, Chen Y, Su Y, Acosta-Baena N, Acosta-Uribe J, Baena AY, Garcia-Ospina G, Giraldo-Chica M, Tirado V, Muñoz C, Ríos-Romenets S, Guzman-Martínez C, Oliveira G, Yang HS, Vila-Castelar C, Pruzin JJ, Ghisays V, Arboleda-Velasquez JF, Kosik KS, Reiman EM, Lopera F, Quiroz YT. Effect of apolipoprotein genotype and educational attainment on cognitive function in autosomal dominant Alzheimer's disease. Nat Commun 2023; 14:5120. [PMID: 37612284 PMCID: PMC10447560 DOI: 10.1038/s41467-023-40775-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 08/09/2023] [Indexed: 08/25/2023] Open
Abstract
Autosomal dominant Alzheimer's disease (ADAD) is genetically determined, but variability in age of symptom onset suggests additional factors may influence cognitive trajectories. Although apolipoprotein E (APOE) genotype and educational attainment both influence dementia onset in sporadic AD, evidence for these effects in ADAD is limited. To investigate the effects of APOE and educational attainment on age-related cognitive trajectories in ADAD, we analyzed data from 675 Presenilin-1 E280A mutation carriers and 594 non-carriers. Here we show that age-related cognitive decline is accelerated in ADAD mutation carriers who also have an APOE e4 allele compared to those who do not and delayed in mutation carriers who also have an APOE e2 allele compared to those who do not. Educational attainment is protective and moderates the effect of APOE on cognition. Despite ADAD mutation carriers being genetically determined to develop dementia, age-related cognitive decline may be influenced by other genetic and environmental factors.
Collapse
Affiliation(s)
| | - N Gil Barksdale
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel Vasquez
- Grupo de Neurociencias de Antioquia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
| | - David Aguillon
- Grupo de Neurociencias de Antioquia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
| | | | - Yi Su
- Banner Alzheimer's Institute, Phoenix, AZ, USA
| | - Natalia Acosta-Baena
- Grupo de Neurociencias de Antioquia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
| | - Juliana Acosta-Uribe
- Grupo de Neurociencias de Antioquia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Ana Y Baena
- Grupo de Neurociencias de Antioquia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
| | - Gloria Garcia-Ospina
- Grupo de Neurociencias de Antioquia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
| | - Margarita Giraldo-Chica
- Grupo de Neurociencias de Antioquia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
| | - Victoria Tirado
- Grupo de Neurociencias de Antioquia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
| | - Claudia Muñoz
- Grupo de Neurociencias de Antioquia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
| | - Silvia Ríos-Romenets
- Grupo de Neurociencias de Antioquia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
| | - Claudia Guzman-Martínez
- Grupo de Neurociencias de Antioquia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
| | - Gabriel Oliveira
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hyun-Sik Yang
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | - Kenneth S Kosik
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, USA
| | | | - Francisco Lopera
- Grupo de Neurociencias de Antioquia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
| | - Yakeel T Quiroz
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Grupo de Neurociencias de Antioquia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia.
| |
Collapse
|
12
|
Sapkota S, Erickson K, Fletcher E, Tomaszewski Farias SE, Jin LW, DeCarli C. Vascular Risk Predicts Plasma Amyloid β 42/40 Through Cerebral Amyloid Burden in Apolipoprotein E ε4 Carriers. Stroke 2023; 54:1227-1235. [PMID: 37021572 PMCID: PMC10121244 DOI: 10.1161/strokeaha.122.041854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/28/2023] [Accepted: 03/08/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND Understanding the neurobiological underpinnings between established multimodal dementia risk factors and noninvasive blood-based biomarkers may lead to greater precision and earlier identification of older adults at risk of accelerated decline and dementia. We examined whether key vascular and genetic risk impact the association between cerebral amyloid burden and plasma aβ (amyloid β) 42/40 in nondemented older adults. METHODS We used nondemented older adults from the UCD-ADRC (University of California, Davis-Alzheimer's Disease Research Center) study (n=96) and Alzheimer's Disease Neuroimaging Initiative (n=104). Alzheimer's Disease Neuroimaging Initiative was examined as confirmatory study cohort. We followed a cross-sectional design and examined linear regression followed by mediation analyses. Vascular risk score was obtained as the sum of hypertension, diabetes, hyperlipidemia, coronary artery disease, and cerebrovascular disease. Apolipoprotein E (APOE) ε4+ risk was genotyped, and plasma aβ42 and aβ40 were assayed. Cerebral amyloid burden was quantified using Florbetapir-PET scans. Baseline age was included as a covariate in all models. RESULTS Vascular risk significantly predicted cerebral amyloid burden in Alzheimer's Disease Neuroimaging Initiative but not in the UCD-ADRC cohort. Cerebral amyloid burden was associated with plasma aβ 42/40 in both cohorts. Higher vascular risk increased cerebral amyloid burden was indirectly associated with reduced plasma aβ 42/40 in Alzheimer's Disease Neuroimaging Initiative but not in UCD-ADRC cohort. However, when stratified by APOE ε4+ risk, we consistently observed this indirect relationship only in APOE ε4+ carriers across both cohorts. CONCLUSIONS Vascular risk is indirectly associated with the level of plasma aβ 42/40 via cerebral amyloid burden only in APOE ε4+ carriers. Nondemented older adults with genetic vulnerability to dementia and accelerated decline may benefit from careful monitoring of vascular risk factors directly associated with cerebral amyloid burden and indirectly with plasma aβ 42/40.
Collapse
Affiliation(s)
- Shraddha Sapkota
- Department of Neurology (S.S., E.F., S.E.T.F., C.D.), University of California, Davis
| | - Kelsey Erickson
- Department of Neurology (S.S., E.F., S.E.T.F., C.D.), University of California, Davis
| | - Evan Fletcher
- University of California, and Department of Pathology and Laboratory Medicine (K.E., L.-W.J.), University of California, Davis
| | | | - Lee-Way Jin
- University of California, and Department of Pathology and Laboratory Medicine (K.E., L.-W.J.), University of California, Davis
| | - Charles DeCarli
- Department of Neurology (S.S., E.F., S.E.T.F., C.D.), University of California, Davis
| |
Collapse
|
13
|
Li X, Kaur Y, Wilhelm O, Reuter M, Montag C, Sommer W, Zhou C, Hildebrandt A. Resting-state brain signal complexity discriminates young healthy APOE e4 carriers from non-e4 carriers. Eur J Neurosci 2023; 57:854-866. [PMID: 36656069 DOI: 10.1111/ejn.15915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 01/20/2023]
Abstract
It is well established that the e4 allele of the APOE gene is associated with impaired brain functionality and cognitive decline in humans at elder age. However, it is controversial whether and how the APOE e4 allele is associated with superior brain function among young healthy individuals, thus indicates a case of antagonistic pleiotropy of APOE e4 allele. Signal complexity is a critical aspect of brain activity that has been associated with brain function. In this study, the multiscale entropy (MSE) of resting-state EEG signals among a sample of young healthy adults (N = 260) as an indicator of brain signal complexity was investigated. It was of interest whether MSE differs across APOE genotype groups while age and education level were controlled for and whether the APOE genotype effect on MSE interacts with MSE time scale, as well as EEG recording condition. Results of linear mixed models indicate overall larger MSE in APOE e4 carriers. This genotype-dependent difference is larger at high as compared with low time scales. The interaction effect between APOE genotype and recording condition indicates increased between-state MSE change in young healthy APOE e4 carriers as compared with non-carriers. Because higher complexity is commonly taken to be associated with better cognitive functioning, the present results complement previous findings and therefore point to a pleiotropic spectrum of the APOE gene polymorphism.
Collapse
Affiliation(s)
- Xiaojing Li
- Chinese Academy of Disability Data Science, Nanjing Normal University of Special Education, Nanjing, China.,Department of Physics, Centre for Nonlinear Studies, Institute of Computational and Theoretical Studies, Hong Kong Baptist University, Hong Kong.,Department of Psychology, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| | - Yadwinder Kaur
- Department of Psychology, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| | | | - Martin Reuter
- Centre for Economics and Neuroscience, University of Bonn, Bonn, Germany.,Department of Psychology, University of Bonn, Bonn, Germany
| | - Christian Montag
- Department of Psychology, Ulm University, Ulm, Germany.,The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, China
| | - Werner Sommer
- Department of Physics, Centre for Nonlinear Studies, Institute of Computational and Theoretical Studies, Hong Kong Baptist University, Hong Kong.,Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany.,Department of Psychology, Zhejiang Normal University, Jinhua, China
| | - Changsong Zhou
- Department of Physics, Centre for Nonlinear Studies, Institute of Computational and Theoretical Studies, Hong Kong Baptist University, Hong Kong
| | - Andrea Hildebrandt
- Department of Psychology, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany.,Research Center Neurosensory Science, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| |
Collapse
|
14
|
Anderson C, Carmichael J, Hicks AJ, Burke R, Ponsford J. Interaction between APOE ɛ4 and Age Is Associated with Emotional Distress One Year after Moderate-Severe Traumatic Brain Injury. J Neurotrauma 2023; 40:326-336. [PMID: 35996348 DOI: 10.1089/neu.2022.0226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Emotional distress is common following moderate-severe traumatic brain injury (TBI) and is associated with poorer post-injury outcomes. Previously investigated sociodemographic, psychological, and injury-related factors account for only a small proportion of variance in post-TBI emotional distress, highlighting a need to consider other factors such as genetic factors. The apolipoprotein E gene (APOE) has been commonly studied in the TBI literature, with the ɛ4 allele linked to worse neuronal repair and recovery. Few studies have investigated the potential relationship between APOE ɛ4 and emotional distress after moderate-severe TBI, and results have been varied. We examined whether APOE ɛ4 was associated with emotional distress 1 year following moderate-severe TBI, and whether this relationship was moderated by age, sex, and TBI severity (as indexed by the duration of post-traumatic amnesia [PTA]). Moderate-severe TBI survivors provided saliva samples following inpatient admission to a TBI rehabilitation hospital. They completed a self-report measure of emotional distress, the Hospital Anxiety and Depression Scale (HADS), at a follow-up interview ∼1 year post-injury. Complete genetic and follow-up data were available for 441 moderate-severe TBI survivors (mean age = 39.42 years; 75% male). We constructed a linear regression model that included APOE ɛ4 carriage status (carrier vs. non-carrier) and interactions with age, sex, and TBI severity (APOE × age, APOE × sex, APOE × age × sex, and APOE × PTA duration) to predict total score on the HADS, while covarying for the main effects of age, sex, PTA duration, and previous head injury. There was a significant main effect of APOE ɛ4, whereby ɛ4 carriers reported less emotional distress than non-carriers (p = 0.04). However, we also found a significant interaction with age such that APOE ɛ4 carriers reported increasingly greater emotional distress with older age compared with non-carriers (p = 0.01). A sensitivity analysis (n = 306) suggested that the APOE × age interaction, and main effects of age and previous head injury, were not unique to individuals with pre-injury mental health problems (n = 136). However, the main effect of APOE ɛ4 was no longer significant when individuals with pre-injury mental health problems were removed. Our findings highlight the importance of considering moderation of genetic associations, suggesting that APOE ɛ4 may be a risk factor for emotional distress specifically among older survivors of moderate-severe TBI. If these findings can be independently replicated, APOE ɛ4 carriage status, interpreted in the context of age, could be incorporated into risk prediction models of emotional distress after moderate-severe TBI, enhancing targeted early detection and intervention efforts.
Collapse
Affiliation(s)
- Chloe Anderson
- Monash-Epworth Rehabilitation Research Centre, Epworth HealthCare, Melbourne, Australia; Turner Institutes for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Victoria, Australia
| | - Jai Carmichael
- Monash-Epworth Rehabilitation Research Centre, Epworth HealthCare, Melbourne, Australia; Turner Institutes for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Victoria, Australia
| | - Amelia J Hicks
- Monash-Epworth Rehabilitation Research Centre, Epworth HealthCare, Melbourne, Australia; Turner Institutes for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Victoria, Australia
| | - Richard Burke
- School of Biological Sciences, Monash University, Clayton, Victoria, Australia
| | - Jennie Ponsford
- Monash-Epworth Rehabilitation Research Centre, Epworth HealthCare, Melbourne, Australia; Turner Institutes for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
15
|
The impact of context on pattern separation for objects among younger and older apolipoprotein ϵ4 carriers and noncarriers. J Int Neuropsychol Soc 2022; 29:439-449. [PMID: 36416211 DOI: 10.1017/s1355617722000455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE On continuous recognition tasks, changing the context objects are embedded in impairs memory. Older adults are worse on pattern separation tasks requiring identification of similar objects compared to younger adults. However, how contexts impact pattern separation in aging is unclear. The apolipoprotein (APOE) ϵ4 allele may exacerbate possible age-related changes due to early, elevated neuropathology. The goal of this study is to determine how context and APOE status affect pattern separation among younger and older adults. METHOD Older and younger ϵ4 carriers and noncarriers were given a continuous object recognition task. Participants indicated if objects on a Repeated White background, Repeated Scene, or a Novel Scene were old, similar, or new. The proportions of correct responses and the types of errors made were calculated. RESULTS Novel scenes lowered recognition scores compared to all other contexts for everyone. Younger adults outperformed older adults on identifying similar objects. Older adults misidentified similar objects as old more than new, and the repeated scene exacerbated this error. APOE status interacted with scene and age such that in repeated scenes, younger carriers produced less false alarms, and this trend switched for older adults where carriers made more false alarms. CONCLUSIONS Context impacted recognition memory in the same way for both age groups. Older adults underutilized details and over relied on holistic information during pattern separation compared to younger adults. The triple interaction in false alarms may indicate an even greater reliance on holistic information among older adults with increased risk for Alzheimer's disease.
Collapse
|
16
|
Steele OG, Stuart AC, Minkley L, Shaw K, Bonnar O, Anderle S, Penn AC, Rusted J, Serpell L, Hall C, King S. A multi-hit hypothesis for an APOE4-dependent pathophysiological state. Eur J Neurosci 2022; 56:5476-5515. [PMID: 35510513 PMCID: PMC9796338 DOI: 10.1111/ejn.15685] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/31/2022] [Accepted: 04/25/2022] [Indexed: 01/01/2023]
Abstract
The APOE gene encoding the Apolipoprotein E protein is the single most significant genetic risk factor for late-onset Alzheimer's disease. The APOE4 genotype confers a significantly increased risk relative to the other two common genotypes APOE3 and APOE2. Intriguingly, APOE4 has been associated with neuropathological and cognitive deficits in the absence of Alzheimer's disease-related amyloid or tau pathology. Here, we review the extensive literature surrounding the impact of APOE genotype on central nervous system dysfunction, focussing on preclinical model systems and comparison of APOE3 and APOE4, given the low global prevalence of APOE2. A multi-hit hypothesis is proposed to explain how APOE4 shifts cerebral physiology towards pathophysiology through interconnected hits. These hits include the following: neurodegeneration, neurovascular dysfunction, neuroinflammation, oxidative stress, endosomal trafficking impairments, lipid and cellular metabolism disruption, impaired calcium homeostasis and altered transcriptional regulation. The hits, individually and in combination, leave the APOE4 brain in a vulnerable state where further cumulative insults will exacerbate degeneration and lead to cognitive deficits in the absence of Alzheimer's disease pathology and also a state in which such pathology may more easily take hold. We conclude that current evidence supports an APOE4 multi-hit hypothesis, which contributes to an APOE4 pathophysiological state. We highlight key areas where further study is required to elucidate the complex interplay between these individual mechanisms and downstream consequences, helping to frame the current landscape of existing APOE-centric literature.
Collapse
Affiliation(s)
| | | | - Lucy Minkley
- School of Life SciencesUniversity of SussexBrightonUK
| | - Kira Shaw
- School of Life SciencesUniversity of SussexBrightonUK
| | - Orla Bonnar
- School of Life SciencesUniversity of SussexBrightonUK
| | | | | | | | | | | | - Sarah King
- School of PsychologyUniversity of SussexBrightonUK
| |
Collapse
|
17
|
Xu J, Guan X, Wen J, Zhang M, Xu X. Polygenic hazard score modified the relationship between hippocampal subfield atrophy and episodic memory in older adults. Front Aging Neurosci 2022; 14:943702. [PMID: 36389062 PMCID: PMC9659745 DOI: 10.3389/fnagi.2022.943702] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 09/30/2022] [Indexed: 12/30/2023] Open
Abstract
BACKGROUND Understanding genetic influences on Alzheimer's disease (AD) may improve early identification. Polygenic hazard score (PHS) is associated with the age of AD onset and cognitive decline. It interacts with other risk factors, but the nature of such combined effects remains poorly understood. MATERIALS AND METHODS We examined the effect of genetic risk and hippocampal atrophy pattern on episodic memory in a sample of older adults ranging from cognitively normal to those diagnosed with AD using structural MRI. Participants included 51 memory unimpaired normal control (NC), 69 mild cognitive impairment (MCI), and 43 AD adults enrolled in the Alzheimer's Disease Neuroimaging Initiative (ADNI). Hierarchical linear regression analyses examined the main and interaction effects of hippocampal subfield volumes and PHS, indicating genetic risk for AD, on a validated episodic memory composite score. Diagnosis-stratified models further assessed the role of PHS. RESULTS Polygenic hazard score moderated the relationship between right fimbria/hippocampus volume ratio and episodic memory, such that patients with high PHS and lower volume ratio had lower episodic memory composite scores [ΔF = 6.730, p = 0.011, ΔR 2 = 0.059]. This effect was also found among individuals with MCI [ΔF = 4.519, p = 0.038, ΔR 2 = 0.050]. In contrast, no interaction effects were present for those NC or AD individuals. A follow-up mediation analysis also indicated that the right fimbria/hippocampus volume ratio might mediate the link between PHS and episodic memory performance in the MCI group, whereas no mediation effects were present for those NC or AD individuals. CONCLUSION These findings suggest that the interaction between AD genetic risk and hippocampal subfield volume ratio increases memory impairment among older adults. Also, the results highlighted a potential pathway in which genetic risk affects memory by degrading hippocampal subfield volume ratio in cognitive decline subjects.
Collapse
Affiliation(s)
| | | | | | | | - Xiaojun Xu
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | | |
Collapse
|
18
|
Shu H, Chen G, Ward BD, Chen G, Wang Z, Liu D, Su F, Gu L, Xu Z, Li SJ, Zhang Z. Imminent cognitive decline in normal elderly individuals is associated with hippocampal hyperconnectivity in the variant neural correlates of episodic memory. Eur Arch Psychiatry Clin Neurosci 2022; 272:783-792. [PMID: 34363508 DOI: 10.1007/s00406-021-01310-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 07/19/2021] [Indexed: 01/06/2023]
Abstract
The secondary prevention trials of Alzheimer's disease (AD) require an enrichment strategy to recruit individuals with imminent cognitive decline at the preclinical stage. Previously, we demonstrated a variant neural correlates of episodic memory (EM) function in apolipoprotein E (APOE) ε4 carriers. Herein, we investigated whether this variation was associated with longitudinal EM performance. This 3-year longitudinal study included 88 normal elderly subjects with EM assessment and resting-state functional MRI data at baseline; 48 subjects (27 ε3 homozygotes and 21 ε4 carriers) underwent follow-up EM assessment. In the identified EM neural correlates, multivariable regression models examined the association between hippocampal functional connectivity (HFC) and longitudinal EM change. Independent validation was performed using the Alzheimer's Disease Neuroimaging Initiative (ADNI) dataset. At baseline, the EM neural correlates were characterized in the Papez circuit regions in the ε3 homozygotes, but in the sensorimotor cortex and cuneus in the ε4 carriers. Longitudinally, the ε4 carriers exhibited a negative association of the baseline HFC strength in the EM neural correlates with annual rate of EM change (R2 = 0.25, p = 0.05). This association also showed a trend in the ADNI dataset (R2 = 0.42, p = 0.06). These results indicate that hippocampal hyperconnectivity in the variant EM neural correlates is associated with imminent EM decline in ε4 carriers, which may serve as a promising enrichment strategy for secondary prevention trials of AD.
Collapse
Affiliation(s)
- Hao Shu
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Neuropsychiatric Institute, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, Jiangsu, China.,Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Gang Chen
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - B Douglas Ward
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Guangyu Chen
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Zan Wang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Neuropsychiatric Institute, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, Jiangsu, China
| | - Duan Liu
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Neuropsychiatric Institute, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, Jiangsu, China
| | - Fan Su
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Neuropsychiatric Institute, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, Jiangsu, China
| | - Lihua Gu
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Neuropsychiatric Institute, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, Jiangsu, China
| | - Zhan Xu
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Shi-Jiang Li
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| | - Zhijun Zhang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Neuropsychiatric Institute, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, Jiangsu, China.
| | | |
Collapse
|
19
|
Diaz JR, Martá-Ariza M, Khodadadi-Jamayran A, Heguy A, Tsirigos A, Pankiewicz JE, Sullivan PM, Sadowski MJ. Apolipoprotein E4 Effects a Distinct Transcriptomic Profile and Dendritic Arbor Characteristics in Hippocampal Neurons Cultured in vitro. Front Aging Neurosci 2022; 14:845291. [PMID: 35572125 PMCID: PMC9099260 DOI: 10.3389/fnagi.2022.845291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 04/06/2022] [Indexed: 11/13/2022] Open
Abstract
The APOE gene is diversified by three alleles ε2, ε3, and ε4 encoding corresponding apolipoprotein (apo) E isoforms. Possession of the ε4 allele is signified by increased risks of age-related cognitive decline, Alzheimer's disease (AD), and the rate of AD dementia progression. ApoE is secreted by astrocytes as high-density lipoprotein-like particles and these are internalized by neurons upon binding to neuron-expressed apoE receptors. ApoE isoforms differentially engage neuronal plasticity through poorly understood mechanisms. We examined here the effects of native apoE lipoproteins produced by immortalized astrocytes homozygous for ε2, ε3, and ε4 alleles on the maturation and the transcriptomic profile of primary hippocampal neurons. Control neurons were grown in the presence of conditioned media from Apoe -/- astrocytes. ApoE2 and apoE3 significantly increase the dendritic arbor branching, the combined neurite length, and the total arbor surface of the hippocampal neurons, while apoE4 fails to produce similar effects and even significantly reduces the combined neurite length compared to the control. ApoE lipoproteins show no systemic effect on dendritic spine density, yet apoE2 and apoE3 increase the mature spines fraction, while apoE4 increases the immature spine fraction. This is associated with opposing effects of apoE2 or apoE3 and apoE4 on the expression of NR1 NMDA receptor subunit and PSD95. There are 1,062 genes differentially expressed across neurons cultured in the presence of apoE lipoproteins compared to the control. KEGG enrichment and gene ontology analyses show apoE2 and apoE3 commonly activate expression of genes involved in neurite branching, and synaptic signaling. In contrast, apoE4 cultured neurons show upregulation of genes related to the glycolipid metabolism, which are involved in dendritic spine turnover, and those which are usually silent in neurons and are related to cell cycle and DNA repair. In conclusion, our work reveals that lipoprotein particles comprised of various apoE isoforms differentially regulate various neuronal arbor characteristics through interaction with neuronal transcriptome. ApoE4 produces a functionally distinct transcriptomic profile, which is associated with attenuated neuronal development. Differential regulation of neuronal transcriptome by apoE isoforms is a newly identified biological mechanism, which has both implication in the development and aging of the CNS.
Collapse
Affiliation(s)
- Jenny R. Diaz
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
| | - Mitchell Martá-Ariza
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
| | | | - Adriana Heguy
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
| | - Aristotelis Tsirigos
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
| | - Joanna E. Pankiewicz
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Biochemistry and Pharmacology, New York University Grossman School of Medicine, New York, NY, United States
| | - Patrick M. Sullivan
- Department of Medicine (Geriatrics), Duke University School of Medicine, Durham, NC, United States
- Durham VA Medical Center’s, Geriatric Research Education and Clinical Center, Durham, NC, United States
| | - Martin J. Sadowski
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Biochemistry and Pharmacology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
20
|
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
|
21
|
Roberts JA, Varma VR, An Y, Varma S, Candia J, Fantoni G, Tiwari V, Anerillas C, Williamson A, Saito A, Loeffler T, Schilcher I, Moaddel R, Khadeer M, Lovett J, Tanaka T, Pletnikova O, Troncoso JC, Bennett DA, Albert MS, Yu K, Niu M, Haroutunian V, Zhang B, Peng J, Croteau DL, Resnick SM, Gorospe M, Bohr VA, Ferrucci L, Thambisetty M. A brain proteomic signature of incipient Alzheimer's disease in young APOE ε4 carriers identifies novel drug targets. SCIENCE ADVANCES 2021; 7:eabi8178. [PMID: 34757788 PMCID: PMC8580310 DOI: 10.1126/sciadv.abi8178] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 09/14/2021] [Indexed: 05/13/2023]
Abstract
Aptamer-based proteomics revealed differentially abundant proteins in Alzheimer’s disease (AD) brains in the Baltimore Longitudinal Study of Aging and Religious Orders Study (mean age, 89 ± 9 years). A subset of these proteins was also differentially abundant in the brains of young APOE ε4 carriers relative to noncarriers (mean age, 39 ± 6 years). Several of these proteins represent targets of approved and experimental drugs for other indications and were validated using orthogonal methods in independent human brain tissue samples as well as in transgenic AD models. Using cell culture–based phenotypic assays, we showed that drugs targeting the cytokine transducer STAT3 and the Src family tyrosine kinases, YES1 and FYN, rescued molecular phenotypes relevant to AD pathogenesis. Our findings may accelerate the development of effective interventions targeting the earliest molecular triggers of AD.
Collapse
Affiliation(s)
- Jackson A. Roberts
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032
| | - Vijay R. Varma
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Yang An
- Brain Aging and Behavior Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | | | - Julián Candia
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Giovanna Fantoni
- Clinical Research Core, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Vinod Tiwari
- Section on DNA Repair, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Carlos Anerillas
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Andrew Williamson
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Atsushi Saito
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Tina Loeffler
- QPS Austria GmbH, Parkring 12, 8074 Grambach, Austria
| | | | - Ruin Moaddel
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Mohammed Khadeer
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Jacqueline Lovett
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Toshiko Tanaka
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Olga Pletnikova
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Juan C. Troncoso
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Marilyn S. Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Kaiwen Yu
- Departments of Structural Biology and Developmental Neurobiology, Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Mingming Niu
- Departments of Structural Biology and Developmental Neurobiology, Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Vahram Haroutunian
- Departments of Psychiatry and Neuroscience, The Alzheimer’s Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mental Illness Research, Education and Clinical Center (MIRECC), James J. Peters VA Medical Center, Bronx, NY 10468, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences and Department of Pharmacological Sciences, Mount Sinai Center for Transformative Disease Modeling, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Junmin Peng
- Departments of Structural Biology and Developmental Neurobiology, Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Deborah L. Croteau
- Section on DNA Repair, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Susan M. Resnick
- Brain Aging and Behavior Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Vilhelm A. Bohr
- Section on DNA Repair, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Madhav Thambisetty
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| |
Collapse
|
22
|
Lu K, Nicholas JM, Pertzov Y, Grogan J, Husain M, Pavisic IM, James SN, Parker TD, Lane CA, Keshavan A, Keuss SE, Buchanan SM, Murray-Smith H, Cash DM, Malone IB, Sudre CH, Coath W, Wong A, Henley SM, Fox NC, Richards M, Schott JM, Crutch SJ. Dissociable effects of APOE-ε4 and β-amyloid pathology on visual working memory. NATURE AGING 2021; 1:1002-1009. [PMID: 34806027 PMCID: PMC7612005 DOI: 10.1038/s43587-021-00117-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 08/17/2021] [Indexed: 01/21/2023]
Abstract
Although APOE-ε4 carriers are at significantly higher risk of developing Alzheimer's disease than non-carriers1, controversial evidence suggests that APOE-ε4 might confer some advantages, explaining the survival of this gene (antagonistic pleiotropy)2,3. In a population-based cohort born in one week in 1946 (assessed aged 69-71), we assessed differential effects of APOE-ε4 and β-amyloid pathology (quantified using 18F-Florbetapir-PET) on visual working memory (object-location binding). In 398 cognitively normal participants, APOE-ε4 and β-amyloid had opposing effects on object identification, predicting better and poorer recall respectively. ε4-carriers also recalled locations more precisely, with a greater advantage at higher β-amyloid burden. These results provide evidence of superior visual working memory in ε4-carriers, showing that some benefits of this genotype are demonstrable in older age, even in the preclinical stages of Alzheimer's disease.
Collapse
Affiliation(s)
- Kirsty Lu
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Jennifer M. Nicholas
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, UK
| | - Yoni Pertzov
- Department of Psychology, The Hebrew University of Jerusalem, Israel
| | - John Grogan
- Nuffield Department of Clinical Neurosciences, University of Oxford, UK
| | - Masud Husain
- Nuffield Department of Clinical Neurosciences, University of Oxford, UK
- Department of Experimental Psychology, University of Oxford, UK
| | - Ivanna M. Pavisic
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, London, UK
| | - Sarah-Naomi James
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, London, UK
| | - Thomas D. Parker
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Christopher A. Lane
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Ashvini Keshavan
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Sarah E. Keuss
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Sarah M. Buchanan
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Heidi Murray-Smith
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - David M. Cash
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute at UCL, University College London, London, UK
| | - Ian B. Malone
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Carole H. Sudre
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, London, UK
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, UK
| | - William Coath
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Andrew Wong
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, London, UK
| | - Susie M.D. Henley
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Nick C. Fox
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute at UCL, University College London, London, UK
| | - Marcus Richards
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, London, UK
| | - Jonathan M. Schott
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Sebastian J. Crutch
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
23
|
Carmichael J, Hicks AJ, Spitz G, Gould KR, Ponsford J. Moderators of gene-outcome associations following traumatic brain injury. Neurosci Biobehav Rev 2021; 130:107-124. [PMID: 34411558 DOI: 10.1016/j.neubiorev.2021.08.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 07/04/2021] [Accepted: 08/13/2021] [Indexed: 12/14/2022]
Abstract
The field of genomics is the principal avenue in the ongoing development of precision/personalised medicine for a variety of health conditions. However, relating genes to outcomes is notoriously complex, especially when considering that other variables can change, or moderate, gene-outcome associations. Here, we comprehensively discuss moderation of gene-outcome associations in the context of traumatic brain injury (TBI), a common, chronically debilitating, and costly neurological condition that is under complex polygenic influence. We focus our narrative review on single nucleotide polymorphisms (SNPs) of three of the most studied genes (apolipoprotein E, brain-derived neurotrophic factor, and catechol-O-methyltransferase) and on three demographic variables believed to moderate associations between these SNPs and TBI outcomes (age, biological sex, and ethnicity). We speculate on the mechanisms which may underlie these moderating effects, drawing widely from biomolecular and behavioural research (n = 175 scientific reports) within the TBI population (n = 72) and other neurological, healthy, ageing, and psychiatric populations (n = 103). We conclude with methodological recommendations for improved exploration of moderators in future genetics research in TBI and other populations.
Collapse
Affiliation(s)
- Jai Carmichael
- Monash-Epworth Rehabilitation Research Centre, Epworth HealthCare, Melbourne, Australia; Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Australia.
| | - Amelia J Hicks
- Monash-Epworth Rehabilitation Research Centre, Epworth HealthCare, Melbourne, Australia; Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Australia
| | - Gershon Spitz
- Monash-Epworth Rehabilitation Research Centre, Epworth HealthCare, Melbourne, Australia; Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Australia
| | - Kate Rachel Gould
- Monash-Epworth Rehabilitation Research Centre, Epworth HealthCare, Melbourne, Australia; Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Australia
| | - Jennie Ponsford
- Monash-Epworth Rehabilitation Research Centre, Epworth HealthCare, Melbourne, Australia; Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Australia
| |
Collapse
|
24
|
Fox-Fuller JT, Artola A, Chen K, Pulsifer M, Ramirez D, Londono N, Aguirre-Acevedo DC, Vila-Castelar C, Baena A, Martinez J, Arboleda-Velasquez JF, Langbaum JB, Tariot PN, Reiman EM, Lopera F, Quiroz YT. Sex Differences in Cognitive Abilities Among Children With the Autosomal Dominant Alzheimer Disease Presenilin 1 E280A Variant From a Colombian Cohort. JAMA Netw Open 2021; 4:e2121697. [PMID: 34463747 PMCID: PMC8408665 DOI: 10.1001/jamanetworkopen.2021.21697] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
IMPORTANCE We previously reported that children with the autosomal dominant Alzheimer disease (ADAD) presenilin 1 (PSEN1) E280A variant had early life plasma biomarker findings consistent with amyloid β overproduction. However, the cognitive functioning of children with this variant has not been characterized vs those without the variant. OBJECTIVE To test whether cognitive functioning of children with and without the PSEN1 E280A variant in the same ADAD cohort differed by genetic status (ie, PSEN1 variant) and sex. DESIGN, SETTING, AND PARTICIPANTS This cohort study was conducted among 1354 children (including 265 children with the variant) aged 6 to 16 years recruited from the Alzheimer Prevention Initiative Colombia Registry. Participants from the city of Medellín and surrounding suburban areas traveled to the University of Antioquia to undergo all procedures. Participants were administered a Spanish version of the Wechsler Intelligence Scale for Children, Fourth Edition (WISC-IV) to measure general cognitive functioning. Data were analyzed from July through November 2020. MAIN OUTCOMES AND MEASURES Univariate general linear models were used to characterize differences on WISC-IV cognitive performance by genetic status, sex, and the interaction of genetic status with sex. Urbanity, socioeconomic status, and education were entered as covariates. RESULTS Among 1354 children with ADAD (695 [51.3%] girls; mean [SD] age, 11.64 [2.64] years), there were 265 children with the variant (19.6%) and 1089 children without the variant (80.4%). Children with and without the variant did not differ by demographic variables or performance on WISC-IV indices. Irrespective of genetic status, boys had statistically significantly decreased mean scores on indices for working memory (90.27 [95% CI, 89.21-91.34] vs 92.99 [95% CI, 91.98-93.99]; mean difference = -2.72; P < .001), perceptual reasoning (91.56 [95% CI, 90.47-92.65] vs. 93.27 [95% CI, 91.23-94.30]; mean difference = -1.71; P = .03), and verbal comprehension (88.69 [95% CI, 87.54-89.84] vs. 90.81 [95% CI, 89.73-91.90]; mean difference = -2.12; P = .009) compared with girls. In the interaction between sex and genetic status, boys with the variant had worse mean working memory index performance (88.78 [95% CI, 86.86-90.70]) than girls with the variant (93.75 [95% CI, 91.95-95.55]; mean difference = -4.97; P = .001), as well as boys (91.77 [95% CI, 90.85-92.70]; mean difference = -2.99; P = .04) and girls (92.22 [95% CI, 91.32-93.13]; mean difference = -3.44; P = .009) without the variant. CONCLUSIONS AND RELEVANCE This study found that boys with the PSEN1 variant had decreased working memory abilities compared with girls with the variant and boys and girls without the variant, suggesting a sex-specific genetic risk in early life cognitive performance among individuals with the PSEN1 variant. This increased risk of future cognitive difficulties among boys with the variant may have important downstream implications for learning and academic achievement and could be associated with sex differences seen in adulthood on episodic memory measures.
Collapse
Affiliation(s)
- Joshua T. Fox-Fuller
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston
- Department of Psychological and Brain Sciences, Boston University, Boston, Massachusetts
| | - Arabiye Artola
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston
- Department of Applied Psychology, Bouvé College of Health Sciences Northeastern University, Boston, Massachusetts
| | - Kewei Chen
- Banner Alzheimer's Institute, Phoenix, Arizona
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe
- Department of Neurology, College of Medicine-Phoenix, University of Arizona, Tempe
| | - Margaret Pulsifer
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Dora Ramirez
- Grupo de Neurociencias, Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Natalia Londono
- Grupo de Neurociencias, Universidad de Antioquia, Medellín, Antioquia, Colombia
| | | | - Clara Vila-Castelar
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Ana Baena
- Grupo de Neurociencias, Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Jairo Martinez
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Joseph F. Arboleda-Velasquez
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston
- Massachusetts Eye and Ear, Harvard Medical School, Boston
| | | | | | | | - Francisco Lopera
- Grupo de Neurociencias, Universidad de Antioquia, Medellín, Antioquia, Colombia
| | - Yakeel T. Quiroz
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston
- Grupo de Neurociencias, Universidad de Antioquia, Medellín, Antioquia, Colombia
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, Massachusetts
- Department of Neurology, Massachusetts General Hospital, Boston
| |
Collapse
|
25
|
Polis B, Karasik D, Samson AO. Alzheimer's disease as a chronic maladaptive polyamine stress response. Aging (Albany NY) 2021; 13:10770-10795. [PMID: 33811757 PMCID: PMC8064158 DOI: 10.18632/aging.202928] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 03/27/2021] [Indexed: 12/21/2022]
Abstract
Polyamines are nitrogen-rich polycationic ubiquitous bioactive molecules with diverse evolutionary-conserved functions. Their activity interferes with numerous genes' expression resulting in cell proliferation and signaling modulation. The intracellular levels of polyamines are precisely controlled by an evolutionary-conserved machinery. Their transient synthesis is induced by heat stress, radiation, and other traumatic stimuli in a process termed the polyamine stress response (PSR). Notably, polyamine levels decline gradually with age; and external supplementation improves lifespan in model organisms. This corresponds to cytoprotective and reactive oxygen species scavenging properties of polyamines. Paradoxically, age-associated neurodegenerative disorders are characterized by upsurge in polyamines levels, indicating polyamine pleiotropic, adaptive, and pathogenic roles. Specifically, arginase overactivation and arginine brain deprivation have been shown to play an important role in Alzheimer's disease (AD) pathogenesis. Here, we assert that a universal short-term PSR associated with acute stimuli is beneficial for survival. However, it becomes detrimental and maladaptive following chronic noxious stimuli, especially in an aging organism. Furthermore, we regard cellular senescence as an adaptive response to stress and suggest that PSR plays a central role in age-related neurodegenerative diseases' pathogenesis. Our perspective on AD proposes an inclusive reassessment of the causal relationships between the classical hallmarks and clinical manifestation. Consequently, we offer a novel treatment strategy predicated upon this view and suggest fine-tuning of arginase activity with natural inhibitors to preclude or halt the development of AD-related dementia.
Collapse
Affiliation(s)
- Baruh Polis
- Drug Discovery Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - David Karasik
- Hebrew SeniorLife, Hinda and Arthur Marcus Institute for Aging Research, Boston, MA 02131, USA
- Musculoskeletal Genetics Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Abraham O. Samson
- Drug Discovery Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| |
Collapse
|
26
|
Iveson MH, Taylor A, Harris SE, Deary IJ, McIntosh AM. Apolipoprotein E e4 allele status and later-life depression in the Lothian Birth Cohort 1936. Psychol Med 2021; 52:1-9. [PMID: 33648619 PMCID: PMC9811345 DOI: 10.1017/s0033291721000623] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 02/03/2021] [Accepted: 02/06/2021] [Indexed: 01/12/2023]
Abstract
BACKGROUND Previous results have been mixed regarding the role of the apolipoprotein E e4 (APOE e4) allele in later-life depression: some studies note that carriers experience greater symptoms and increased risk while others find no such association. However, there are few prospective, population-based studies of the APOE e4-depression association and fewer that examine depressive symptom trajectory and depression risk longitudinally. We examined the association between APOE e4 allele status and longitudinal change in depressive symptoms and depression risk in later-life, over a 12-year follow-up period. METHODS We used data from 690 participants of the Lothian Birth Cohort 1936 who took part in the Scottish Mental Survey 1947 (aged 11) and were followed-up in later-life over five waves from 2004 to 2019 (aged 70-82). We used APOE e4 allele status to predict longitudinal change in depressive symptom scores and risk of depression (defined by a symptom score threshold or use of depression-related medication). Models were adjusted for sex, childhood cognitive ability, childhood social class, education, adult social class, smoking status and functional limitations at baseline. RESULTS Depressive symptom scores increased with age. Once adjusted for covariates, APOE e4 allele status did not significantly predict symptom score trajectories or depression risk. Greater functional limitations at baseline significantly predicted poorer symptom score trajectories and increased depression risk (defined by medications). APOE e4 allele status did not significantly moderate the contribution of sex, education or functional limitations. CONCLUSIONS There was no evidence that APOE e4 carriers experience an increased risk for later-life depression.
Collapse
Affiliation(s)
- Matthew H. Iveson
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
- Mental Health Data Science Scotland, Edinburgh, UK
| | - Adele Taylor
- Lothian Birth Cohorts, The University of Edinburgh, Edinburgh, UK
| | - Sarah E. Harris
- Lothian Birth Cohorts, The University of Edinburgh, Edinburgh, UK
| | - Ian J. Deary
- Lothian Birth Cohorts, The University of Edinburgh, Edinburgh, UK
| | - Andrew M. McIntosh
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, UK
- Mental Health Data Science Scotland, Edinburgh, UK
| |
Collapse
|
27
|
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: 0.7] [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
|
28
|
Sanabria-Diaz G, Melie-Garcia L, Draganski B, Demonet JF, Kherif F. Apolipoprotein E4 effects on topological brain network organization in mild cognitive impairment. Sci Rep 2021; 11:845. [PMID: 33436948 PMCID: PMC7804004 DOI: 10.1038/s41598-020-80909-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 12/30/2020] [Indexed: 01/29/2023] Open
Abstract
The Apolipoprotein E isoform E4 (ApoE4) is consistently associated with an elevated risk of developing late-onset Alzheimer's Disease (AD); however, less is known about the potential genetic modulation of the brain networks organization during prodromal stages like Mild Cognitive Impairment (MCI). To investigate this issue during this critical stage, we used a dataset with a cross-sectional sample of 253 MCI patients divided into ApoE4-positive (‛Carriers') and ApoE4-negative ('non-Carriers'). We estimated the cortical thickness (CT) from high-resolution T1-weighted structural magnetic images to calculate the correlation among anatomical regions across subjects and build the CT covariance networks (CT-Nets). The topological properties of CT-Nets were described through the graph theory approach. Specifically, our results showed a significant decrease in characteristic path length, clustering-index, local efficiency, global connectivity, modularity, and increased global efficiency for Carriers compared to non-Carriers. Overall, we found that ApoE4 in MCI shaped the topological organization of CT-Nets. Our results suggest that in the MCI stage, the ApoE4 disrupting the CT correlation between regions may be due to adaptive mechanisms to sustain the information transmission across distant brain regions to maintain the cognitive and behavioral abilities before the occurrence of the most severe symptoms.
Collapse
Affiliation(s)
- Gretel Sanabria-Diaz
- Laboratoire de Recherche en Neuroimagerie (LREN), Département des neurosciences cliniques, Centre Hospitalier Universitaire Vaudois (CHUV), Mont Paisible 16, 1011, Lausanne, Switzerland.
| | - Lester Melie-Garcia
- Laboratoire de Recherche en Neuroimagerie (LREN), Département des neurosciences cliniques, Centre Hospitalier Universitaire Vaudois (CHUV), Mont Paisible 16, 1011, Lausanne, Switzerland
| | - Bogdan Draganski
- Laboratoire de Recherche en Neuroimagerie (LREN), Département des neurosciences cliniques, Centre Hospitalier Universitaire Vaudois (CHUV), Mont Paisible 16, 1011, Lausanne, Switzerland
| | | | - Ferath Kherif
- Laboratoire de Recherche en Neuroimagerie (LREN), Département des neurosciences cliniques, Centre Hospitalier Universitaire Vaudois (CHUV), Mont Paisible 16, 1011, Lausanne, Switzerland
| |
Collapse
|
29
|
Hays CC, Zlatar ZZ, Meloy MJ, Bondi MW, Gilbert PE, Liu T, Helm JL, Wierenga CE. Interaction of APOE, cerebral blood flow, and cortical thickness in the entorhinal cortex predicts memory decline. Brain Imaging Behav 2021; 14:369-382. [PMID: 32048144 DOI: 10.1007/s11682-019-00245-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The ε4 allele of the apolipoprotein E (APOE) gene, a risk factor for cognitive decline, is associated with alterations in medial temporal lobe (MTL) structure and function, yet little research has been dedicated to understanding how these alterations might interact to negatively impact cognition. To bridge this gap, the present study employed linear regression models to determine the extent to which APOE genotype (ε4+, ε4-) modifies interactive effects of baseline arterial spin labeling MRI-measured cerebral blood flow (CBF) and FreeSurfer-derived cortical thickness/volume (CT/Vo) in two MTL regions of interest (entorhinal cortex, hippocampus) on memory change in 98 older adults who were cognitively normal at baseline. Baseline entorhinal CBF was positively associated with memory change, but only among ε4 carriers with lower entorhinal CT. Similarly, baseline entorhinal CT was positively associated with memory change, but only among ε4 carriers with lower entorhinal CBF. Findings suggest that APOE ε4 carriers may experience concomitant alterations in neurovascular function and morphology in the MTL that interact to negatively affect cognition prior to the onset of overt clinical symptoms. Results also suggest the presence of distinct multimodal neural signatures in the entorhinal cortex that may signal relative risk for cognitive decline among this group, perhaps reflecting different stages of cerebrovascular compensation (early effective vs. later ineffective).
Collapse
Affiliation(s)
- Chelsea C Hays
- VA San Diego Healthcare System, 3350 La Jolla Village Dr., MC 151B, San Diego, CA, 9216, USA.,SDSU/UCSD Joint Doctoral Program in Clinical Psychology, 6363 Alvarado Ct, Suite 103, San Diego, CA, 92120, USA
| | - Zvinka Z Zlatar
- Department of Psychiatry, UC San Diego, 9500 Gilman Dr., La Jolla, CA, 92093, USA
| | - M J Meloy
- VA San Diego Healthcare System, 3350 La Jolla Village Dr., MC 151B, San Diego, CA, 9216, USA.,Department of Psychiatry, UC San Diego, 9500 Gilman Dr., La Jolla, CA, 92093, USA
| | - Mark W Bondi
- VA San Diego Healthcare System, 3350 La Jolla Village Dr., MC 151B, San Diego, CA, 9216, USA.,SDSU/UCSD Joint Doctoral Program in Clinical Psychology, 6363 Alvarado Ct, Suite 103, San Diego, CA, 92120, USA.,Department of Psychiatry, UC San Diego, 9500 Gilman Dr., La Jolla, CA, 92093, USA
| | - Paul E Gilbert
- SDSU/UCSD Joint Doctoral Program in Clinical Psychology, 6363 Alvarado Ct, Suite 103, San Diego, CA, 92120, USA.,Department of Psychology, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Thomas Liu
- Department of Radiology, UC San Diego, 9500 Gilman Dr., La Jolla, CA, 92093, USA
| | - Jonathan L Helm
- SDSU/UCSD Joint Doctoral Program in Clinical Psychology, 6363 Alvarado Ct, Suite 103, San Diego, CA, 92120, USA.,Department of Psychology, San Diego State University, 5500 Campanile Drive, San Diego, CA, 92182, USA
| | - Christina E Wierenga
- VA San Diego Healthcare System, 3350 La Jolla Village Dr., MC 151B, San Diego, CA, 9216, USA. .,SDSU/UCSD Joint Doctoral Program in Clinical Psychology, 6363 Alvarado Ct, Suite 103, San Diego, CA, 92120, USA. .,Department of Psychiatry, UC San Diego, 9500 Gilman Dr., La Jolla, CA, 92093, USA.
| |
Collapse
|
30
|
Gharbi-Meliani A, Dugravot A, Sabia S, Regy M, Fayosse A, Schnitzler A, Kivimäki M, Singh-Manoux A, Dumurgier J. The association of APOE ε4 with cognitive function over the adult life course and incidence of dementia: 20 years follow-up of the Whitehall II study. ALZHEIMERS RESEARCH & THERAPY 2021; 13:5. [PMID: 33397450 PMCID: PMC7784268 DOI: 10.1186/s13195-020-00740-0] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 12/07/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Approximately 25% of the general population carries at least one ε4 allele of the Apolipoprotein E (APOE ε4), the strongest genetic risk factor for late onset Alzheimer's disease. Beyond its association with late-onset dementia, the association between APOE ε4 and change in cognition over the adult life course remains uncertain. This study aims to examine whether the association between Apolipoprotein E (APOE) ε4 zygosity and cognition function is modified between midlife and old age. METHODS A cohort study of 5561 participants (mean age 55.5 (SD = 5.9) years, 27.1% women) with APOE genotyping and repeated cognitive tests for reasoning, memory, and semantic and phonemic fluency, during a mean (SD) follow-up of 20.2 (2.8) years (the Whitehall II study). We used joint models to examine the association of APOE genotype with cognitive function trajectories between 45 and 85 years taking drop-out, dementia, and death into account and Fine and Gray models to examine associations with dementia. RESULTS Compared to non-carriers, heterozygote (prevalence 25%) and homozygote (prevalence 2%) APOE ε4 carriers had increased risk of dementia, sub-distribution hazard ratios 2.19 (95% CI 1.73, 2.77) and 5.97 (95% CI 3.85, 9.28) respectively. Using data spanning 45-85 years with non-ε4 carriers as the reference, ε4 homozygotes had poorer global cognitive score starting from 65 years; ε4 heterozygotes had better scores between 45 and 55 years, then no difference until poorer cognitive scores from 75 years onwards. In analysis of individual cognitive tests, better cognitive performance in the younger ε4 heterozygotes was primarily attributable to executive function. CONCLUSIONS Both heterozygous and homozygous ε4 carriers had poorer cognition and greater risk of dementia at older ages. Our findings show some support for a complex antagonist pleiotropic effect of APOE ε4 heterozygosity over the adult life course, characterized by cognitive advantage in midlife.
Collapse
Affiliation(s)
- Amin Gharbi-Meliani
- Université de Paris, Inserm U1153, Epidemiology of Ageing and Neurodegenerative Diseases, Paris, France
| | - Aline Dugravot
- Université de Paris, Inserm U1153, Epidemiology of Ageing and Neurodegenerative Diseases, Paris, France
| | - Séverine Sabia
- Université de Paris, Inserm U1153, Epidemiology of Ageing and Neurodegenerative Diseases, Paris, France
| | - Melina Regy
- Université de Paris, Inserm U1153, Epidemiology of Ageing and Neurodegenerative Diseases, Paris, France
| | - Aurore Fayosse
- Université de Paris, Inserm U1153, Epidemiology of Ageing and Neurodegenerative Diseases, Paris, France
| | - Alexis Schnitzler
- Université de Paris, Inserm U1153, Epidemiology of Ageing and Neurodegenerative Diseases, Paris, France
| | - Mika Kivimäki
- Department of Epidemiology and Public Health, University College London, London, UK
| | - Archana Singh-Manoux
- Université de Paris, Inserm U1153, Epidemiology of Ageing and Neurodegenerative Diseases, Paris, France.,Department of Epidemiology and Public Health, University College London, London, UK
| | - Julien Dumurgier
- Université de Paris, Inserm U1153, Epidemiology of Ageing and Neurodegenerative Diseases, Paris, France. .,Cognitive Neurology Center, Lariboisiere - Fernand Widal Hospital, AP-HP, Université de Paris, 200 rue du Faubourg Saint Denis, 75010, Paris, France.
| |
Collapse
|
31
|
Martí-Juan G, Sanroma-Guell G, Cacciaglia R, Falcon C, Operto G, Molinuevo JL, González Ballester MÁ, Gispert JD, Piella G. Nonlinear interaction between APOE ε4 allele load and age in the hippocampal surface of cognitively intact individuals. Hum Brain Mapp 2020; 42:47-64. [PMID: 33017488 PMCID: PMC7721244 DOI: 10.1002/hbm.25202] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/16/2020] [Accepted: 08/11/2020] [Indexed: 01/27/2023] Open
Abstract
The ε4 allele of the gene Apolipoprotein E is the major genetic risk factor for Alzheimer's Disease. APOE ε4 has been associated with changes in brain structure in cognitively impaired and unimpaired subjects, including atrophy of the hippocampus, which is one of the brain structures that is early affected by AD. In this work we analyzed the impact of APOE ε4 gene dose and its association with age, on hippocampal shape assessed with multivariate surface analysis, in a ε4‐enriched cohort of n = 479 cognitively healthy individuals. Furthermore, we sought to replicate our findings on an independent dataset of n = 969 individuals covering the entire AD spectrum. We segmented the hippocampus of the subjects with a multi‐atlas‐based approach, obtaining high‐dimensional meshes that can be analyzed in a multivariate way. We analyzed the effects of different factors including APOE, sex, and age (in both cohorts) as well as clinical diagnosis on the local 3D hippocampal surface changes. We found specific regions on the hippocampal surface where the effect is modulated by significant APOE ε4 linear and quadratic interactions with age. We compared between APOE and diagnosis effects from both cohorts, finding similarities between APOE ε4 and AD effects on specific regions, and suggesting that age may modulate the effect of APOE ε4 and AD in a similar way.
Collapse
Affiliation(s)
- Gerard Martí-Juan
- BCN MedTech, Departament de Tecnologies de la Informació i les Comunicacions, Universitat Pompeu Fabra, Barcelona, Spain
| | | | - Raffaele Cacciaglia
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Carles Falcon
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBERBBN), Madrid, Spain
| | - Grégory Operto
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| | - Miguel Ángel González Ballester
- BCN MedTech, Departament de Tecnologies de la Informació i les Comunicacions, Universitat Pompeu Fabra, Barcelona, Spain.,ICREA, Barcelona, Spain
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center (BBRC), Pasqual Maragall Foundation, Barcelona, Spain.,Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBERBBN), Madrid, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| | - Gemma Piella
- BCN MedTech, Departament de Tecnologies de la Informació i les Comunicacions, Universitat Pompeu Fabra, Barcelona, Spain
| | | | | |
Collapse
|
32
|
Trdin A, Snoj Tratnik J, Stajnko A, Marc J, Mazej D, Sešek Briški A, Kastelec D, Prpić I, Petrović O, Špirić Z, Horvat M, Falnoga I. Trace elements and APOE polymorphisms in pregnant women and their new-borns. ENVIRONMENT INTERNATIONAL 2020; 143:105626. [PMID: 32622117 DOI: 10.1016/j.envint.2020.105626] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 02/28/2020] [Accepted: 02/29/2020] [Indexed: 06/11/2023]
Abstract
We investigated the relationship between lipid binding glycoprotein apolipoprotein E (apoE; gene APOE) polymorphisms (ε4 allele carriers versus no carriers = ε4+/ε4-) and trace elements (TEs) (e.g., (methyl)mercury, arsenic, lead, cadmium, selenium, manganese, copper, and zinc) in mothers (N = 223) and their new-borns (N = 213) exposed to potentially toxic metal(loid)s from seafood consumption. The apoE isoform encoded by the ε4 allele is believed to have beneficial effects in early life but represents a risk factor for age-associated diseases. Under certain conditions ε4 carriers are more susceptible to oxidative stress and metal(loid) toxicity. DNA from Croatian pregnant women (N = 223, third trimester) and their new-borns (N = 176), was genotyped for APOE by TaqMan® SNP assay - rs429358 and rs7412. Seafood intake data and TE levels in maternal urine, milk, hair, peripheral venous blood, mixed cord blood, and new-borns' urine were available from previous studies. We compared TEs between ε4+ and ε4- carriers using Mann-Whitney U tests and applied multiple linear regression models to analyse the TE's dependence on the presence of allele ε4 (genotypes ε3/ε4, ε4/ε4) in combination with other explanatory variables. We identified 17% (n = 37) and 20% (n = 35) ε4 allele carriers in mothers and new-borns, respectively. The Mann-Whitney U test showed that mothers with the ε4 allele had significantly higher mean levels of (methyl)mercury in peripheral venous blood, cord blood, and hair; arsenic in urine and cord blood; and selenium in peripheral venous blood and plasma. However, taking confounders into account, only the maternal plasma selenium remained statistically significant in the linear regression models (ε4 carriers vs non-carriers: 62.6 vs 54.9 ng/mL, p < 0.001). Literature suggestions of possible ε4 allele impact on Hg levels were not observed, while superior selenium status observed in healthy pregnant women carrying allele ε4 could be linked to the proposed APOE ε4 beneficial effects early in life.
Collapse
Affiliation(s)
- Ajda Trdin
- Department of Environmental Sciences, Jožef Stefan Institute, Ljubljana, Slovenia; Jožef Stefan International Postgraduate School, Ljubljana, Slovenia
| | - Janja Snoj Tratnik
- Department of Environmental Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Anja Stajnko
- Department of Environmental Sciences, Jožef Stefan Institute, Ljubljana, Slovenia; Jožef Stefan International Postgraduate School, Ljubljana, Slovenia
| | - Janja Marc
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Darja Mazej
- Department of Environmental Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Alenka Sešek Briški
- Institute of Clinical Chemistry and Biochemistry, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | | | - Igor Prpić
- Department of Paediatrics, University Hospital Centre Rijeka, Rijeka, Croatia; Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Oleg Petrović
- Department of Paediatrics, University Hospital Centre Rijeka, Rijeka, Croatia
| | | | - Milena Horvat
- Department of Environmental Sciences, Jožef Stefan Institute, Ljubljana, Slovenia; Jožef Stefan International Postgraduate School, Ljubljana, Slovenia
| | - Ingrid Falnoga
- Department of Environmental Sciences, Jožef Stefan Institute, Ljubljana, Slovenia.
| |
Collapse
|
33
|
Smith CJ, Ashford JW, Perfetti TA. Putative Survival Advantages in Young Apolipoprotein ɛ4 Carriers are Associated with Increased Neural Stress. J Alzheimers Dis 2020; 68:885-923. [PMID: 30814349 PMCID: PMC6484250 DOI: 10.3233/jad-181089] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Inheritance of a single copy of the apolipoprotein E (APOE) ɛ4 allele increases risk of Alzheimer’s disease (AD) by 3-4-fold, with homozygosity associated with a 12-16-fold increase in risk, relative to ɛ3 allele homozygosity. There is a decreased risk associated with the APOE ɛ2 allele. The pathological consequence of APOE genotype has led to intense efforts to understand the mechanistic basis of the interplay between APOE status and loss of synapses. Numerous ɛ4 allele-related associations have been reported with the potential relevance of these associations to the pathogenesis of AD unknown at this time. In primarily young subjects, we have reviewed a representative body of literature on ɛ4 allele-associations related to the following: cardiovascular responses; impacts on reproduction and fetal development; co-morbidities; resistance to infectious disease; responses to head injury; biochemical differences possibly related to neural stress; and brain structure-function differences. In addition, the literature on the association between the ɛ4 allele and cognitive performance has been reviewed comprehensively. The weight-of-the-evidence supports the hypothesis that possession of the ancestral ɛ4 allele in youth is associated with improved fitness during fetal development, infancy, and youth relative to the more recently appearing ɛ3 allele, at the expense of decreased fitness in old age, which is substantially improved by the ɛ3 allele. However, possession of the ɛ4 allele is also associated with higher levels of synaptic macromolecular turnover, which likely stresses basic cellular neuroplasticity mechanisms. Clinical trials of potential AD therapeutics should consider APOE status as an enrollment criterion.
Collapse
Affiliation(s)
- Carr J Smith
- Florida State University, Department of Nurse Anesthesia, Panama City, FL, USA
| | - J Wesson Ashford
- Stanford University and VA Palo Alto Health Care System, Palo Alto, CA, USA
| | | |
Collapse
|
34
|
Dzianok P, Kublik E. Commentary: Differential Signaling Mediated by ApoE2, ApoE3, and ApoE4 in Human Neurons Parallels Alzheimer's Disease Risk. Front Aging Neurosci 2020; 12:127. [PMID: 32457598 PMCID: PMC7225266 DOI: 10.3389/fnagi.2020.00127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 04/15/2020] [Indexed: 01/19/2023] Open
Affiliation(s)
- Patrycja Dzianok
- Laboratory of Emotions Neurobiology, Nencki Institute of Experimental Biology PAS, Warsaw, Poland
| | - Ewa Kublik
- Laboratory of Emotions Neurobiology, Nencki Institute of Experimental Biology PAS, Warsaw, Poland
| |
Collapse
|
35
|
Burns RA, Andrews S, Cherbuin N, Anstey KJ. Role of apolipoprotein E epsilon 4 ( APOE*ε4) as an independent risk factor for incident depression over a 12-year period in cognitively intact adults across the lifespan. BJPsych Open 2020; 6:e47. [PMID: 32381152 PMCID: PMC7331079 DOI: 10.1192/bjo.2020.29] [Citation(s) in RCA: 8] [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] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The apolipoprotein E ε4 allele (APOE*ε4) is indicated as a risk for Alzheimer's disease and other age-related diseases. The risk attributable to APOE*ε4 for depression is less clear and may be because of confounding of the relationship between dementia and depression. AIMS We examined the risk of APOE* ε4 for incident depression and depressive symptomology over a 12-year period across the adult lifespan. METHOD Participants were from the Personality and Total Health Through Life study, aged 20 to 24 (n = 1420), 40 to 44 (n = 1592) or 60-64 (n = 1768) at baseline, and interviewed every 4 years since 1999. Ethnicities other than White, those without genotyping and those with depression at baseline, or who reported strokes and scores on the Mini-Mental State Examination <27 at any observation, were excluded. RESULTS Over the study period, there was no evidence that APOE*ε4+ was a risk factor for depression, including any depression (odds ratio (OR) = 0.94, 95% CI 0.77-1.16, P = 0.573), major depression (OR = 0.96, 95% CI 0.60-1.53, P = 0.860), minor depression (OR = 0.94, 95% CI 0.67-1.30, P = 0.695) or depressive symptomology (incidence rate ratio (IRR) = 1.02, 95% CI 0.97-1.08, P = 0.451). APOE*ε4 was unrelated to incident depression. Findings were consistent for all age cohorts. CONCLUSIONS Among cognitively intact Australian adults who were free of depression at baseline, there was little evidence that APOE*ε4+ carriers are at increased risk for depression over a 12-year period among those who are cognitively intact.
Collapse
Affiliation(s)
- Richard Andrew Burns
- Centre for Research on Ageing, Health and Wellbeing, The Australian National University, Australia
| | - Shea Andrews
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, USA
| | - Nicolas Cherbuin
- Centre for Research on Ageing, Health and Wellbeing, The Australian National University, Australia
| | - Kaarin Jane Anstey
- School of Psychology, University of New South Wales, Australia; and Neuroscience Research Australia, Australia
| |
Collapse
|
36
|
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: 3.5] [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
|
37
|
Jeon SY, Byun MS, Yi D, Lee JH, Ko K, Sohn BK, Lee JY, Ryu SH, Lee DW, Shin SA, Kim YK, Kang KM, Sohn CH, Lee DY. Midlife Lifestyle Activities Moderate APOE ε4 Effect on in vivo Alzheimer's Disease Pathologies. Front Aging Neurosci 2020; 12:42. [PMID: 32256335 PMCID: PMC7093017 DOI: 10.3389/fnagi.2020.00042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 02/07/2020] [Indexed: 12/24/2022] Open
Abstract
This study aimed to investigate whether the midlife cognitive activity and physical activity moderate the relationship between apolipoprotein Eε4 (APOE4) and in vivo Alzheimer's disease (AD) pathologies. In total, 287 non-demented older adults (mean age 72 years) from the Korean Brain Aging Study for the Early diagnosis and prediction of Alzheimer's disease cohort were included. Participants underwent a comprehensive clinical assessment including the evaluation for midlife CA and physical activity, [11C]-Pittsburgh-Compound-B-positron emission tomography (PET), [18F]-fluorodeoxyglucose PET, structural magnetic resonance imaging (MRI), and APOE genotyping. We used linear regression and regression-based mediated-moderation models for statistical analyses. Neither midlife cognitive activity nor physical activity moderated the effect of APOE4 on β-amyloid (Aβ) retention itself. Midlife cognitive activity significantly moderated the effect of APOE4 on hippocampal volume [B (SE) = - 627.580 (252.327), t = -2.488, p = 0.014]: APOE4 carriers had smaller hippocampal volume than non-carriers at relatively high cognitive activity state (p = 0.004), but not at relatively low cognitive activity condition (p = 0.937). Midlife physical activity significantly moderated the effect of Aβ retention, which was closely related to APOE4, on AD-signature region cerebral glucose metabolism [AD-CM; B (SE) = 0.004 (0.002), t = 2.030, p = 0.043]: higher Aβ accumulation was associated with lower AD-CM in relatively low physical activity condition (p < 0.001), whereas no such association was observed in relatively high physical activity state (p = 0.791). The findings suggest that high midlife cognitive activity may accelerate hippocampal atrophy induced by APOE4, whereas high midlife physical activity may delay AD-related cerebral hypometabolism by weakening the influence of APOE4-associated Aβ retention.
Collapse
Affiliation(s)
- So Yeon Jeon
- Department of Psychiatry, Chungnam National University Hospital, Daejeon, South Korea
| | - Min Soo Byun
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, South Korea
| | - Jun-Ho Lee
- Department of Psychiatry, National Center for Mental Health, Seoul, South Korea
| | - Kang Ko
- Department of Psychiatry, National Center for Mental Health, Seoul, South Korea
| | - Bo Kyung Sohn
- Department of Neuropsychiatry, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, South Korea
| | - Jun-Young Lee
- Department of Neuropsychiatry, SMG-SNU Boramae Medical Center, Seoul, South Korea
| | - Seung-Ho Ryu
- Department of Psychiatry, School of Medicine, Konkuk University Medical Center, Konkuk University, Seoul, South Korea
| | - Dong Woo Lee
- Department of Neuropsychiatry, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, South Korea
| | - Seoung A Shin
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, South Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, South Korea
| | - Koung Mi Kang
- Department of Radiology, Seoul National University Hospital, Seoul, South Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, Seoul, South Korea
| | - Dong Young Lee
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea.,Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, South Korea.,Department of Psychiatry, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
38
|
Bråthen ACS, De Lange AMG, Fjell AM, Walhovd KB. Risk- and protective factors for memory plasticity in aging. AGING NEUROPSYCHOLOGY AND COGNITION 2020; 28:201-217. [PMID: 32098566 DOI: 10.1080/13825585.2020.1727834] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Risk and protective factors for cognitive function in aging may affect how much individuals benefit from their environment or life experiences by preserving or improving cognitive abilities. We investigated the relations between such factors and outcome from episodic-memory training in 136 healthy young and older adults. Tested risk factors included carrying the ɛ4 variant of the apolipoprotein E allele (APOE), age, body mass index, blood pressure, and cholesterol. Protective factors included higher levels of education, intelligence quotient (IQ), physical activity, fatty acids, and vitamin D. Average increases in memory performance were seen after training, with ample variation between individuals. Being young, female, and having higher IQ were positive predictors of memory improvement. No other relationships were observed. Similar benefit was observed across APOE allelic variation. This indicates that beyond IQ, age, and sex, known risk -and protective factors of cognitive function in aging were not significantly related to memory plasticity.
Collapse
Affiliation(s)
- Anne Cecilie Sjøli Bråthen
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo , Oslo, Norway
| | - Ann-Marie Glasø De Lange
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo , Oslo, Norway
| | - Anders M Fjell
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo , Oslo, Norway.,Department of Radiology and Nuclear Medicine, Oslo University Hospital , Oslo, Norway
| | - Kristine B Walhovd
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo , Oslo, Norway.,Department of Radiology and Nuclear Medicine, Oslo University Hospital , Oslo, Norway
| |
Collapse
|
39
|
Mid age APOE ε4 carriers show memory-related functional differences and disrupted structure-function relationships in hippocampal regions. Sci Rep 2020; 10:3110. [PMID: 32080211 PMCID: PMC7033211 DOI: 10.1038/s41598-020-59272-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 01/22/2020] [Indexed: 01/05/2023] Open
Abstract
Carriers of the APOE e4 allele are at higher risk of age-related cognitive decline and Alzheimer’s disease (AD). The underlying neural mechanisms are uncertain, but genotype differences in medial temporal lobe (MTL) functional activity and structure at mid-age might contribute. We tested 16 non-e4 and 16 e4 carriers (aged 45–55) on a subsequent memory task in conjunction with MRI to assess how hippocampal volume (from T1 structural) and microstructure (neurite orientation-dispersion, from NODDI) differs by genotype and in relation to memory encoding. No previous study has investigated APOE effects on hippocampal microstructure using NODDI. Recall performance did not differ by genotype. A genotype by condition interaction in left parahippocampus indicated that in e4 carriers activity did not differentiate subsequently remembered from forgotten words. Hippocampal volumes and microstructure also did not differ by genotype but hippocampal volumes correlated positively with recognition performance in non-e4 carriers only. Similarly, greater hippocampal neurite orientation-dispersion was linked to better recall but only in non-e4s. Thus, we suggest that mid-age e4 carriers show a breakdown of normal MTL activation and structure-performance relationships. This could reflect an inability to utilise compensatory mechanisms, and contribute to higher risk of cognitive decline and AD in later life.
Collapse
|
40
|
Beyond the CNS: The many peripheral roles of APOE. Neurobiol Dis 2020; 138:104809. [PMID: 32087284 DOI: 10.1016/j.nbd.2020.104809] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 02/06/2020] [Accepted: 02/18/2020] [Indexed: 12/28/2022] Open
Abstract
Apolipoprotein E (APOE) is a multifunctional protein synthesized and secreted by multiple mammalian tissues. Although hepatocytes contribute about 75% of the peripheral pool, APOE can also be expressed in adipose tissue, the kidney, and the adrenal glands, among other tissues. High levels of APOE production also occur in the brain, where it is primarily synthesized by glia, and peripheral and brain APOE pools are thought to be distinct. In humans, APOE is polymorphic, with three major alleles (ε2, ε3, and ε4). These allelic forms dramatically alter APOE structure and function. Historically, the vast majority of research on APOE has centered on the important role it plays in modulating risk for cardiovascular disease and Alzheimer's disease. However, the established effects of this pleiotropic protein extend well beyond these two critical health challenges, with demonstrated roles across a wide spectrum of biological conditions, including adipose tissue function and obesity, metabolic syndrome and diabetes, fertility and longevity, and immune function. While the spectrum of biological systems in which APOE plays a role seems implausibly wide at first glance, there are some potential unifying mechanisms that could tie these seemingly disparate disorders together. In the current review, we aim to concisely summarize a wide breadth of APOE-associated pathologies and to analyze the influence of APOE in the development of several distinct disorders in order to provide insight into potential shared mechanisms implied in these various pathophysiological processes.
Collapse
|
41
|
APOE alters glucose flux through central carbon pathways in astrocytes. Neurobiol Dis 2020; 136:104742. [PMID: 31931141 DOI: 10.1016/j.nbd.2020.104742] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/04/2020] [Accepted: 01/08/2020] [Indexed: 02/02/2023] Open
Abstract
The Apolipoprotein E (APOE) gene is a major genetic risk factor associated with Alzheimer's disease (AD). APOE encodes for three main isoforms in humans (E2, E3, and E4). Homozygous E4 individuals have more than a 10-fold higher risk for developing late-onset AD, while E2 carriers are protected. A hallmark of AD is a reduction in cerebral glucose metabolism, alluding to a strong metabolic component in disease onset and progression. Interestingly, E4 individuals display a similar regional pattern of cerebral glucose hypometabolism decades prior to disease onset. Mapping this metabolic landscape may help elucidate the underlying biological mechanism of APOE-associated risk for AD. Efficient metabolic coupling of neurons and glia is necessary for proper neuronal function, and disruption in glial energy distribution has been proposed to contribute to neuronal cell death and AD pathology. One important function of astrocytes - canonically the primary source of apolipoprotein E in the brain - is to provide metabolic substrates (lactate, lipids, amino acids and neurotransmitters) to neurons. Here we investigate the effects of APOE on astrocyte glucose metabolism in vitro utilizing scintillation proximity assays, stable isotope tracer metabolomics, and gene expression analyses. Glucose uptake is impaired in E4 astrocytes relative to E2 or E3 with specific alterations in central carbon metabolism. Using stable isotope labeled glucose [U-13C] allowed analyses of astrocyte-specific deep metabolic networks affected by APOE, and provided insight to the effects downstream of glucose uptake. Enrichment of 13C in early steps of glycolysis was lowest in E4 astrocytes (highest in E2), while synthesis of lactate from glucose was highest in E4 astrocytes (lowest in E2). We observed an increase in glucose flux through the pentose phosphate pathway (PPP), with downstream increases in gluconeogenesis, lipid, and de novo nucleotide biosynthesis in E4 astrocytes. There was also a marked increase in 13C enrichment in the TCA cycle of E4 astrocytes - whose substrates were also incorporated into biosynthetic pathways at a higher rate. Pyruvate carboxylase (PC) and pyruvate dehydrogenase (PDH) are the two main enzymes controlling pyruvate entry to the TCA cycle. PC gene expression is increased in E4 astrocytes and the activity relative to PDH was also increased, compared to E2 or E3. Decreased enrichment in the TCA cycle of E2 and E3 astrocytes is suggestive of increased oxidation and non-glucose derived anaplerosis, which could be fueling mitochondrial ATP production. Conversely, E4 astrocytes appear to increase carbon flux into the TCA cycle to fuel cataplerosis. Together, these data demonstrate clear APOE isoform-specific effects on glucose utilization in astrocytes, including E4-associated increases in lactate synthesis, PPP flux, and de novo biosynthesis pathways.
Collapse
|
42
|
Rabipour S, Rajagopal S, Yu E, Pasvanis S, Lafaille-Magnan ME, Breitner J, Rajah MN. APOE4 Status is Related to Differences in Memory-Related Brain Function in Asymptomatic Older Adults with Family History of Alzheimer's Disease: Baseline Analysis of the PREVENT-AD Task Functional MRI Dataset. J Alzheimers Dis 2020; 76:97-119. [PMID: 32474466 PMCID: PMC7369116 DOI: 10.3233/jad-191292] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND Episodic memory decline is one of the earliest symptoms of late-onset Alzheimer's disease (AD). Older adults with the apolipoprotein E ɛ4 (+APOE4) genetic risk factor for AD may exhibit altered patterns of memory-related brain activity years prior to initial symptom onset. OBJECTIVE Here we report the baseline episodic memory task functional MRI results from the PRe-symptomatic EValuation of Experimental or Novel Treatments for Alzheimer's Disease cohort in Montreal, Canada, in which 327 healthy older adults were scanned within 15 years of their parent's conversion to AD. METHODS Volunteers were scanned as they encoded and retrieved object-location spatial source associations. The task was designed to discriminate between brain activity related to spatial source recollection and object-only (recognition) memory. We used multivariate partial least squares (PLS) to test the hypothesis that +APOE4 adults with family history of AD would exhibit altered patterns of brain activity in the recollection-related memory network, comprised of medial frontal, parietal, and medial temporal cortices, compared to APOE4 non-carriers (-APOE4). We also examined group differences in the correlation between event-related brain activity and memory performance. RESULTS We found group similarities in memory performance and in task-related brain activity in the recollection network, but differences in brain activity-behavior correlations in ventral occipito-temporal, medial temporal, and medial prefrontal cortices during episodic encoding. CONCLUSION These findings are consistent with previous literature on the influence of APOE4 on brain activity and provide new perspective on potential gene-based differences in brain-behavior relationships in people with first-degree family history of AD.
Collapse
Affiliation(s)
- Sheida Rabipour
- Centre for Cerebral Imaging, Douglas Hospital Research Centre, Montreal, Canada
- Department of Psychiatry, McGill University, Montreal, Canada
| | | | - Elsa Yu
- Integrated Program in Neuroscience, McGill University, Montreal, Canada
| | - Stamatoula Pasvanis
- Centre for Cerebral Imaging, Douglas Hospital Research Centre, Montreal, Canada
| | - Marie-Elyse Lafaille-Magnan
- Department of Psychiatry, McGill University, Montreal, Canada
- Center for Studies on Prevention of Alzheimer’s Disease, Montreal, Canada
- Lady Davis Center for Medical Research, Jewish General Hospital, Montreal, Canada
| | - John Breitner
- Centre for Cerebral Imaging, Douglas Hospital Research Centre, Montreal, Canada
- Center for Studies on Prevention of Alzheimer’s Disease, Montreal, Canada
| | | | - M. Natasha Rajah
- Centre for Cerebral Imaging, Douglas Hospital Research Centre, Montreal, Canada
- Department of Psychiatry, McGill University, Montreal, Canada
| |
Collapse
|
43
|
Hashimoto M, Ho G, Takamatsu Y, Shimizu Y, Sugama S, Takenouchi T, Waragai M, Masliah E. Evolvability and Neurodegenerative Disease: Antagonistic Pleiotropy Phenomena Derived from Amyloid Aggregates. JOURNAL OF PARKINSONS DISEASE 2019; 8:405-408. [PMID: 30010144 PMCID: PMC6130413 DOI: 10.3233/jpd-181365] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
At present, the precise physiological role of neurodegenerative disease-related amyloidogenic proteins (APs), including α-synuclein in Parkinson’s disease and β-amyloid in Alzheimer’s disease, remains unclear. Because of similar adaptability of both human brain neurons and yeast cells to diverse environmental stressors, we previously proposed that the concept of evolvability in yeast prion could also be applied to APs in human brain. However, the mechanistic relevance of evolvability to neurodegenerative disorders is elusive. Therefore, our objective is to discuss our hypothesis that evolvability and neurodegenerative disease may represent a form of antagonistic pleiotropy derived from the aggregates of APs. Importantly, such a perspective may provide an outlook of the entire course of sporadic neurodegenerative diseases.
Collapse
Affiliation(s)
- Makoto Hashimoto
- Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Gilbert Ho
- PCND Neuroscience Research Institute, Poway, CA, USA
| | - Yoshiki Takamatsu
- Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Yuka Shimizu
- Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Shuei Sugama
- Department of Physiology, Nippon Medical School, Tokyo, Japan
| | - Takato Takenouchi
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
| | - Masaaki Waragai
- Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Eliezer Masliah
- Division of Neurosciences, National Institute on Aging, Bethesda, MD, USA
| |
Collapse
|
44
|
Trdin A, Snoj Tratnik J, Mazej D, Fajon V, Krsnik M, Osredkar J, Prpić I, Špirić Z, Petrović O, Marc J, Neubauer D, Kodrič J, Kobal AB, Barbone F, Falnoga I, Horvat M. Mercury speciation in prenatal exposure in Slovenian and Croatian population - PHIME study. ENVIRONMENTAL RESEARCH 2019; 177:108627. [PMID: 31421448 DOI: 10.1016/j.envres.2019.108627] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 07/30/2019] [Accepted: 08/04/2019] [Indexed: 06/10/2023]
Abstract
In recent years, several studies have addressed the issue of prenatal exposure to methylmercury (MeHg); however, few have actually analysed MeHg blood concentrations. Our study population included mothers and their new-borns from Slovenia (central region; N = 584) and Croatia (coastal region; N = 234). We have measurements of total Hg (THg) and MeHg in maternal hair, maternal peripheral blood, and cord blood. Cord blood Hg concentrations were low to moderate (median THg = 1.84 ng/g and MeHg = 1.69 ng/g). The proportion of THg as MeHg (%MeHg) in maternal and cord blood varied between 4% and 100% (coefficient of variation, CV = 32%) and between 8% and 100% (CV = 20%), respectively. Our data shows that variability of %MeHg was higher at lower blood THg levels. Concentrations of MeHg in maternal blood and cord blood were highly correlated (Rs = 0.943), in the case of inorganic Hg correlation was significant but weaker (Rs = 0.198). MeHg levels in maternal blood and cord blood were positively associated with seafood intake, maternal age, and negatively associated with pre-pregnancy BMI. Additionally, MeHg in maternal blood was positively associated with plasma selenium levels, and cord blood MeHg was negatively associated with parity. The results of multiple linear regression models showed that speciation analysis provides more defined estimation of prenatal exposure in association modelling. Associations between Hg exposure and cognitive performance of children (assessed using Bayley Scales of Infant and Toddler development) adjusted for maternal or child Apolipoprotein E genotypes showed higher model R2 and lower p-values when adjusted for MeHg compared to THg. This study demonstrates that Hg speciation improves the association between exposure and possible negative health effects.
Collapse
Affiliation(s)
- Ajda Trdin
- Department of Environmental Sciences, Jožef Stefan Institute, Ljubljana, Slovenia; Jožef Stefan International Postgraduate School, Ljubljana, Slovenia
| | - Janja Snoj Tratnik
- Department of Environmental Sciences, Jožef Stefan Institute, Ljubljana, Slovenia; Jožef Stefan International Postgraduate School, Ljubljana, Slovenia
| | - Darja Mazej
- Department of Environmental Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Vesna Fajon
- Department of Environmental Sciences, Jožef Stefan Institute, Ljubljana, Slovenia; Jožef Stefan International Postgraduate School, Ljubljana, Slovenia
| | - Mladen Krsnik
- Institute of Clinical Chemistry and Biochemistry, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Joško Osredkar
- Institute of Clinical Chemistry and Biochemistry, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Igor Prpić
- Department of Paediatrics, University Hospital Centre Rijeka, Rijeka, Croatia
| | | | - Oleg Petrović
- Department of Obstetrics and Gynaecology, University Hospital Centre Rijeka, Rijeka, Croatia
| | - Janja Marc
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - David Neubauer
- Department of Child, Adolescent, and Developmental Neurology, Division of Paediatrics, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Jana Kodrič
- Department of Child, Adolescent, and Developmental Neurology, Division of Paediatrics, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Alfred B Kobal
- Department of Environmental Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Fabio Barbone
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | - Ingrid Falnoga
- Department of Environmental Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Milena Horvat
- Department of Environmental Sciences, Jožef Stefan Institute, Ljubljana, Slovenia; Jožef Stefan International Postgraduate School, Ljubljana, Slovenia.
| |
Collapse
|
45
|
APOE modifies the interaction of entorhinal cerebral blood flow and cortical thickness on memory function in cognitively normal older adults. Neuroimage 2019; 202:116162. [PMID: 31493534 DOI: 10.1016/j.neuroimage.2019.116162] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 08/14/2019] [Accepted: 09/03/2019] [Indexed: 11/20/2022] Open
Abstract
OBJECTIVE The ε4 allele of the apolipoprotein E (APOE) gene increases risk for cognitive decline in normal and pathologic aging. However, precisely how APOE ε4 exerts its negative impact on cognition is poorly understood. The present study aimed to determine whether APOE genotype (ε4+ vs. ε4-) modifies the interaction of medial temporal lobe (MTL) resting cerebral blood flow (CBF) and brain structure (cortical thickness [CT], volume [Vo]) on verbal memory performance. METHODS Multiple linear regression models were employed to investigate relationships between APOE genotype, arterial spin labeling MRI-measured CBF and FreeSurfer-based CT and Vo in four MTL regions of interest (left and right entorhinal cortex and hippocampus), and verbal memory performance among a sample of 117 cognitively normal older adults (41 ε4+, 76 ε4-) between the ages of 64 and 89 (mean age = 73). RESULTS Results indicated that APOE genotype modified the interaction of CBF and CT on memory in the left entorhinal cortex, such that the relationship between entorhinal CBF and memory was negative (lower CBF was associated with better memory) in non-carriers with higher entorhinal CT, positive (higher CBF was associated with better memory) in non-carriers with lower entorhinal CT, and negative (higher CBF was associated with worse memory) in ε4 carriers with lower entorhinal CT. CONCLUSIONS Findings suggest that older adult APOE ε4 carriers may experience vascular dysregulation and concomitant morphological alterations in the MTL that interact to negatively affect memory even in the absence overt clinical symptoms, providing potential insight into the mechanistic link between APOE ε4 and detriments in cognition. Moreover, findings suggest a distinct multimodal neural signature in ε4 carriers (higher CBF and lower CT in the entorhinal cortex) that could aid in the identification of candidates for future clinical trials aimed at preventing or slowing cognitive decline. Differential findings with respect to ε4 carriers and non-carriers are discussed in the context of neurovascular compensation.
Collapse
|
46
|
Korthauer LE, Awe E, Frahmand M, Driscoll I. Genetic Risk for Age-Related Cognitive Impairment Does Not Predict Cognitive Performance in Middle Age. J Alzheimers Dis 2019; 64:459-471. [PMID: 29865048 DOI: 10.3233/jad-171043] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Alzheimer's disease (AD) is characterized by memory loss and executive dysfunction, which correspond to structural changes to the medial temporal lobes (MTL) and prefrontal cortex (PFC), respectively. Given the overlap in cognitive deficits between healthy aging and the earliest stages of AD, early detection of AD remains a challenge. The goal of the present study was to study MTL- and PFC-dependent cognitive functioning in middle-aged individuals at genetic risk for AD or cognitive impairment who do not currently manifest any clinical symptoms. Participants (N = 150; aged 40-60 years) underwent genotyping of 47 single nucleotide polymorphisms (SNPs) in six genes previously associated with memory or executive functioning: APOE, SORL1, BDNF, TOMM40, KIBRA, and COMT. They completed two MTL-dependent tasks, the virtual Morris Water Task (vMWT) and transverse patterning discriminations task (TPDT), and the PFC-dependent reversal learning task. Although age was associated with poorer performance on the vMWT and TPDT within this middle-aged sample, there were no genotype-associated differences in cognitive performance. Although the vMWT and TPDT may be sensitive to age-related changes in cognition, carriers of APOE, SORL1, BDNF, TOMM40, KIBRA, and COMT risk alleles do not exhibit alteration in MTL- and PFC-dependent functioning in middle age compared to non-carriers.
Collapse
Affiliation(s)
- Laura E Korthauer
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Elizabeth Awe
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA.,Department of Neurology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Marijam Frahmand
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Ira Driscoll
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| |
Collapse
|
47
|
Shu H, Shi Y, Chen G, Wang Z, Liu D, Yue C, Ward BD, Li W, Xu Z, Chen G, Guo QH, Xu J, Li SJ, Zhang Z. Distinct neural correlates of episodic memory among apolipoprotein E alleles in cognitively normal elderly. Brain Imaging Behav 2019; 13:255-269. [PMID: 29396739 DOI: 10.1007/s11682-017-9818-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The apolipoprotein E (APOE) ε4 and ε2 alleles are acknowledged genetic factors modulating Alzheimer's disease (AD) risk and episodic memory (EM) deterioration in an opposite manner. Mounting neuroimaging studies describe EM-related brain activity differences among APOE alleles but remain limited in elucidating the underlying mechanism. Here, we hypothesized that the APOE ε2, ε3, and ε4 alleles have distinct EM neural substrates, as a manifestation of degeneracy, underlying their modulations on EM-related brain activity and AD susceptibility. To test the hypothesis, we identified neural correlates of EM function by correlating intrinsic hippocampal functional connectivity networks with neuropsychological EM performances in a voxelwise manner, with 129 cognitively normal elderly subjects (36 ε2 carriers, 44 ε3 homozygotes, and 49 ε4 carriers). We demonstrated significantly different EM neural correlates among the three APOE allele groups. Specifically, in the ε3 homozygotes, positive EM neural correlates were characterized in the Papez circuit regions; in the ε4 carriers, positive EM neural correlates involved the lateral temporal cortex, premotor cortex/sensorimotor cortex/superior parietal lobule, and cuneus; and in the ε2 carriers, negative EM neural correlates appeared in the bilateral frontopolar, posteromedial, and sensorimotor cortex. Further, in the ε4 carriers, the interaction between age and EM function occurred in the temporoparietal junction and prefrontal cortex. Our findings suggest that the underlying mechanism of APOE polymorphism modulations on EM function and AD susceptibility is genetically related to the neural degeneracy of EM function across APOE alleles.
Collapse
Affiliation(s)
- Hao Shu
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, Jiangsu, 210009, China
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Yongmei Shi
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, Jiangsu, 210009, China
| | - Gang Chen
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Zan Wang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, Jiangsu, 210009, China
| | - Duan Liu
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, Jiangsu, 210009, China
| | - Chunxian Yue
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, Jiangsu, 210009, China
| | - B Douglas Ward
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Wenjun Li
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Zhan Xu
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Guangyu Chen
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Qi-Hao Guo
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jun Xu
- Department of Neurology, Jiangsu Province Geriatric Institute, Nanjing, Jiangsu, 210024, China
| | - Shi-Jiang Li
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| | - Zhijun Zhang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, Jiangsu, 210009, China.
| |
Collapse
|
48
|
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: 14] [Impact Index Per Article: 2.8] [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
|
49
|
Female mice with apolipoprotein E4 domain interaction demonstrated impairments in spatial learning and memory performance and disruption of hippocampal cyto-architecture. Neurobiol Learn Mem 2019; 161:106-114. [PMID: 30954674 DOI: 10.1016/j.nlm.2019.03.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 03/01/2019] [Accepted: 03/29/2019] [Indexed: 11/22/2022]
Abstract
We have previously reported cognitive impairments in both young and old mice, particularly in female mice expressing mouse Arg-61 apoE, with a point mutation to mimic the domain interaction feature of human apoE4, as compared to the wildtype mouse (C57BL/6J) apoE. In this study, we further evaluated water maze performance in the female Arg-61 mice at an additional time point and then investigated related hippocampal cyto-architecture in these young female Arg-61 apoE mice vs. the wildtype mice. The results of behavioral performance consistently support our previous report that the young female Arg-61 apoE showed cognitive impairment versus C57BL/6J at the same age. The cyto-architectural results showed that volume of the granular cell layer (GCL) was significantly larger in both 5- and 10-month old Arg-61 apoE mice versus C57BL/6J mice. While the number of newborn calretinin-positive neurons was greater in the sub-granular zone (SGZ) in 5-month old Arg-61 mice, this number dropped significantly in 10-month old Arg-61 mice to a lower level than in age-matched C57BL/6J mice. In addition, the amyloid β species was significantly higher in 5-month old Arg-61 mice versus age-matched C57BL/6J mice. In conclusion, impaired cognitive functions in female Arg-61 apoE mice appear correlated with larger GCL volume and higher calretinin-positive cell number and suggest a compensatory cellular response that may be related to amyloid beta perturbations early in life. Therefore this study suggests a novel cyto-architectural mechanism of apoE4-dependent pathologies and increased susceptibility of APOEε4 subjects to Alzheimer's disease.
Collapse
|
50
|
The Contribution of Genetic Factors to Cognitive Impairment and Dementia: Apolipoprotein E Gene, Gene Interactions, and Polygenic Risk. Int J Mol Sci 2019; 20:ijms20051177. [PMID: 30866553 PMCID: PMC6429136 DOI: 10.3390/ijms20051177] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 02/26/2019] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease. Although it has been studied for years, the pathogenesis of AD is still controversial. Genetic factors may play an important role in pathogenesis, with the apolipoprotein E (APOE) gene among the greatest risk factors for AD. In this review, we focus on the influence of genetic factors, including the APOE gene, the interaction between APOE and other genes, and the polygenic risk factors for cognitive function and dementia. The presence of the APOE ε4 allele is associated with increased AD risk and reduced age of AD onset. Accelerated cognitive decline and abnormal internal environment, structure, and function of the brain were also found in ε4 carriers. The effect of the APOE promoter on cognition and the brain was confirmed by some studies, but further investigation is still needed. We also describe the effects of the associations between APOE and other genetic risk factors on cognition and the brain that exhibit a complex gene⁻gene interaction, and we consider the importance of using a polygenic risk score to investigate the association between genetic variance and phenotype.
Collapse
|