151
|
Bruña R, Maestú F, López-Sanz D, Bagic A, Cohen AD, Chang YF, Cheng Y, Doman J, Huppert T, Kim T, Roush RE, Snitz BE, Becker JT. Sex Differences in Magnetoencephalography-Identified Functional Connectivity in the Human Connectome Project Connectomics of Brain Aging and Dementia Cohort. Brain Connect 2021; 12:561-570. [PMID: 34726478 PMCID: PMC9419974 DOI: 10.1089/brain.2021.0059] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Introduction: The human brain shows modest traits of sexual dimorphism, with the female brain, on average, 10% smaller than the male brain. These differences do not imply a lowered cognitive performance, but suggest a more optimal brain organization in women. Here we evaluate the patterns of functional connectivity (FC) in women and men from the Connectomics of Brain Aging and Dementia sample. Methods: We used phase locking values to calculate FC from the magnetoencephalography time series in a sample of 138 old adults (87 females and 51 males). We compared the FC patterns between sexes, with the intention of detecting regions with different levels of connectivity. Results: We found a frontal cluster, involving anterior cingulate and the medial frontal lobe, where women showed higher FC values than men. Involved connections included the following: (1) medial parietal areas, such as posterior cingulate cortices and precunei; (2) right insula; and (3) medium cingulate and paracingulate cortices. Moreover, these differences persisted when considering only cognitively intact individuals, but not when considering only cognitively impaired individuals. Discussion: Increased anteroposterior FC has been identified as a biomarker for increased risk of developing cognitive impairment or dementia. In our study, cognitively intact women showed higher levels of FC than their male counterparts. This result suggests that neurodegenerative processes could be taking place in these women, but the changes are undetected by current diagnosis tools. FC, as measured here, might be valuable for early identification of this neurodegeneration.
Collapse
Affiliation(s)
- Ricardo Bruña
- Laboratory of Cognitive and Computational Neuroscience (UCM-UPM), Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain.,Department of Experimental Psychology, Universidad Complutense de Madrid, Pozuelo de Alarcón, Madrid, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Fernando Maestú
- Laboratory of Cognitive and Computational Neuroscience (UCM-UPM), Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain.,Department of Experimental Psychology, Universidad Complutense de Madrid, Pozuelo de Alarcón, Madrid, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - David López-Sanz
- Laboratory of Cognitive and Computational Neuroscience (UCM-UPM), Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain.,Department of Psychobiology, Universidad Complutense de Madrid, Madrid, Spain
| | - Anto Bagic
- Department of Psychiatry, The University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Statistics, The University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ann D Cohen
- Department of Neurosurgery, The University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yue-Fang Chang
- Department of Neurosurgery, The University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yu Cheng
- Department of Statistics, The University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Biostatistics, The University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jack Doman
- Department of Neurosurgery, The University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ted Huppert
- Department of Electrical Engineering, The University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Tae Kim
- Department of Radiology, The University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rebecca E Roush
- Department of Psychiatry, The University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Beth E Snitz
- Department of Psychiatry, The University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - James T Becker
- Department of Psychiatry, The University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Neurology, and The University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Psychology, The University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
152
|
Udeh-Momoh C, Watermeyer T. Female specific risk factors for the development of Alzheimer's disease neuropathology and cognitive impairment: Call for a precision medicine approach. Ageing Res Rev 2021; 71:101459. [PMID: 34508876 DOI: 10.1016/j.arr.2021.101459] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 08/13/2021] [Accepted: 09/06/2021] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) includes a long asymptomatic stage, which precedes the formal diagnosis of dementia. AD biomarker models provide a framework for precision medicine approaches during this stage. However, such approaches have ignored the possible influence of sex on cognition and brain health, despite female sex noted as a major risk factor. Since AD-related changes may emerge in midlife, intervention efforts are being redirected around this period. Midlife coincides with several endocrinological changes, such as the menopausal transition experienced by women. In this narrative review, we discuss evidence for sex-differences in AD neuropathological burden and outline key endocrinological mechanisms for both sexes, focussing on hormonal events throughout the lifespan that may influence female susceptibility to AD neuropathology and dementia onset. We further consider common non-modifiable (genetic) and modifiable (lifestyle and health) risk factors, highlighting possible sex-dependent differential effects for the AD disease course. Finally, we evaluate the studies selected for this review demonstrating sex-differences in cognitive, pathological and health factors, summarising the state of sex differences in AD risk factors. We further provide recommendations for targeted research on female-specific risk factors, to inform personalised strategies for AD-prevention and the promotion of female brain health.
Collapse
|
153
|
Kim H, Jun S, Kim BS, Kim IJ. Serum Adiponectin in Alzheimer's Disease (AD): Association with AD Biomarkers and Cognitive Outcome. J Alzheimers Dis 2021; 84:1163-1172. [PMID: 34633322 DOI: 10.3233/jad-210722] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND The association between dementia and serum adiponectin has been evaluated in many studies; however, conclusions remain mixed. OBJECTIVE We investigated the cross-sectional associations of adiponectin with cognitive function and Alzheimer's disease (AD) biomarkers and whether serum adiponectin levels can predict cognitive outcomes. METHODS This study included 496 participants from the Alzheimer's Disease Neuroimaging Initiative 1 (ADNI1) with available serum adiponectin levels at baseline and ≥65 years of age. Subjects were stratified based on sex and apolipoprotein ɛ4 (APOE4) carrier status to determine associations between adiponectin and cognitive function. The linear mixed model was used to analyze associations between adiponectin level and cognitive outcome in amnestic mild cognitive impairment (aMCI) patients. RESULTS Serum adiponectin levels were higher in aMCI and AD than in CN subjects among APOE4 non-carrier males (adiponectin in CN, aMCI, and AD: 0.54±0.24, 0.74±0.25, and 0.85±0.25, respectively, p < 0.001). In this group, serum adiponectin levels were associated with age (p = 0.001), ADAS13 (p < 0.001), memory function (p < 0.001), executive function (p < 0.001), total tau (p < 0.001), and phosphorylated tau (p < 0.001) measures in cerebrospinal fluid (CSF). Higher adiponectin level was not associated with cognitive outcome in aMCI patients in the linear mixed model analysis over 5.3±2.6 years of mean follow-up. CONCLUSION Serum adiponectin level was associated with cognitive function and CSF AD biomarkers among APOE4 non-carrier males. However, serum adiponectin level was not associated with longitudinal cognitive function outcome in aMCI.
Collapse
Affiliation(s)
- Heeyoung Kim
- Departement of Nuclear Medicine, Kosin University Gospel Hospital, University of Kosin College of Medicine, Busan, Republic of Korea
| | - Sungmin Jun
- Departement of Nuclear Medicine, Kosin University Gospel Hospital, University of Kosin College of Medicine, Busan, Republic of Korea
| | - Bum Soo Kim
- Departement of Nuclear Medicine, Kosin University Gospel Hospital, University of Kosin College of Medicine, Busan, Republic of Korea
| | - In-Joo Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Pusan National University School of Medicine, Busan, Republic of Korea
| | | |
Collapse
|
154
|
Papazoglou A, Arshaad MI, Henseler C, Daubner J, Broich K, Haenisch B, Weiergräber M. The Janus-like Association between Proton Pump Inhibitors and Dementia. Curr Alzheimer Res 2021; 18:453-469. [PMID: 34587884 PMCID: PMC8778640 DOI: 10.2174/1567205018666210929144740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 06/10/2021] [Accepted: 06/18/2021] [Indexed: 11/30/2022]
Abstract
Early pharmacoepidemiological studies suggested that Proton Pump Inhibitors (PPIs) might increase the risk of Alzheimer’s Disease (AD) and non-AD related dementias. These findings were supported by preclinical studies, specifically stressing the proamyloidogenic and indirect anticholinergic effects of PPIs. However, further large-scale pharmacoepidemiological studies showed inconsistent results on the association between PPIs and dementia. Pharmacodynamically, these findings might be related to the LXR/RXR-mediated amyloid clearance effect and anti-inflammatory action of PPIs. Further aspects that influence PPI effects on AD are related to patient-specific pharmacokinetic and pharmacogenomic characteristics. In conclusion, a personalized (individualized) medicinal approach is necessary to model and predict the potential harmful or beneficial effects of PPIs in AD and non-AD-related dementias in the future.
Collapse
Affiliation(s)
- Anna Papazoglou
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Muhammad I Arshaad
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Christina Henseler
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Johanna Daubner
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Karl Broich
- Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM) 53127, Kurt-Georg- Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Britta Haenisch
- Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM) 53127, Kurt-Georg- Kiesinger-Allee 3, 53175 Bonn, Germany
| | - Marco Weiergräber
- Experimental Neuropsychopharmacology, Federal Institute for Drugs and Medical Devices (Bundesinstitut für Arzneimittel und Medizinprodukte, BfArM), Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| |
Collapse
|
155
|
Sriwichaiin S, Chattipakorn N, Chattipakorn SC. Metabolomic Alterations in the Blood and Brain in Association with Alzheimer's Disease: Evidence from in vivo to Clinical Studies. J Alzheimers Dis 2021; 84:23-50. [PMID: 34511504 DOI: 10.3233/jad-210737] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Alzheimer's disease (AD) has become a major health problem among the elderly population. Some evidence suggests that metabolic disturbance possibly plays a role in the pathophysiology of AD. Currently, the study of metabolomics has been used to explore changes in multiple metabolites in several diseases, including AD. Thus, the metabolomics research in AD might provide some information regarding metabolic dysregulations, and their possible associated pathophysiology. This review summarizes the information discovered regarding the metabolites in the brain and the blood from the metabolomics research of AD from both animal and clinical studies. Additionally, the correlation between the changes in metabolites and outcomes, such as pathological findings in the brain and cognitive impairment are discussed. We also deliberate on the findings of cohort studies, demonstrating the alterations in metabolites before changes of cognitive function. All of these findings can be used to inform the potential identity of specific metabolites as possible biomarkers for AD.
Collapse
Affiliation(s)
- Sirawit Sriwichaiin
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand.,Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
156
|
Schwartz JB, Weintraub S. Treatment for Alzheimer Disease-Sex and Gender Effects Need to Be Explicitly Analyzed and Reported in Clinical Trials. JAMA Netw Open 2021; 4:e2124386. [PMID: 34515788 DOI: 10.1001/jamanetworkopen.2021.24386] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Janice B Schwartz
- Department of Medicine, University of California, San Francisco
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco
| | - Sandra Weintraub
- Department ofPsychiatry and Behavioral Science, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
157
|
Li M, Lin J, Liang S, Chen Z, Bai Y, Long X, Huang S, Mo Z. The role of age at menarche and age at menopause in Alzheimer's disease: evidence from a bidirectional mendelian randomization study. Aging (Albany NY) 2021; 13:19722-19749. [PMID: 34347623 PMCID: PMC8386554 DOI: 10.18632/aging.203384] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 05/31/2021] [Indexed: 12/16/2022]
Abstract
The association between endogenous estrogen exposure and Alzheimer's disease (AD) remains inconclusive in previous observational studies, and few Mendelian randomization (MR) studies have focused on their causality thus far. We performed a bidirectional MR study to clarify the causality and causal direction of age at menarche and age at menopause, which are indicators of endogenous estrogen exposure, on AD risk. We obtained all genetic datasets for the MR analyses using publicly available summary statistics based on individuals of European ancestry from the IEU GWAS database. The MR analyses indicated no significant causal relationship between the genetically determined age at menarche (outlier-adjusted inverse variance weighted odds ratio [IVWOR] = 0.926; 95% confidence interval [CI], 0.803-1.066) or age at menopause (outlier-adjusted IVWOR = 0.981; 95% CI, 0.941-1.022) and AD risk. Similarly, AD did not show any causal association with age at menarche or age at menopause. The sensitivity analyses yielded similar results. In contrast, an inverse association was detected between age at menarche and body mass index (BMI, outlier-adjusted IVW β = -0.043; 95% CI, -0.077 to -0.009). Our bidirectional MR study provides no evidence for a causal relationship between the genetically determined age at menarche or age at menopause and AD susceptibility, or vice versa. However, earlier menarche might be associated with higher adult BMI.
Collapse
Affiliation(s)
- Mingli Li
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning 530021, Guangxi, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Nanning 530021, Guangxi, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning 530021, Guangxi, China.,Guangxi Key Laboratory of Colleges and Universities, Nanning 530021, Guangxi, China.,School of Public Health of Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Jiali Lin
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning 530021, Guangxi, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Nanning 530021, Guangxi, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning 530021, Guangxi, China.,Guangxi Key Laboratory of Colleges and Universities, Nanning 530021, Guangxi, China.,School of Public Health of Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Shuang Liang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning 530021, Guangxi, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Nanning 530021, Guangxi, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning 530021, Guangxi, China.,Guangxi Key Laboratory of Colleges and Universities, Nanning 530021, Guangxi, China.,School of Public Health of Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Zefeng Chen
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning 530021, Guangxi, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Nanning 530021, Guangxi, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning 530021, Guangxi, China.,Guangxi Key Laboratory of Colleges and Universities, Nanning 530021, Guangxi, China.,School of Public Health of Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Yulan Bai
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning 530021, Guangxi, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Nanning 530021, Guangxi, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning 530021, Guangxi, China.,Guangxi Key Laboratory of Colleges and Universities, Nanning 530021, Guangxi, China.,School of Public Health of Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Xinyang Long
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning 530021, Guangxi, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Nanning 530021, Guangxi, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning 530021, Guangxi, China.,Guangxi Key Laboratory of Colleges and Universities, Nanning 530021, Guangxi, China.,School of Public Health of Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Shengzhu Huang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning 530021, Guangxi, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Nanning 530021, Guangxi, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning 530021, Guangxi, China.,Guangxi Key Laboratory of Colleges and Universities, Nanning 530021, Guangxi, China.,School of Public Health of Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Zengnan Mo
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning 530021, Guangxi, China.,Guangxi Key Laboratory for Genomic and Personalized Medicine, Nanning 530021, Guangxi, China.,Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Nanning 530021, Guangxi, China.,Guangxi Key Laboratory of Colleges and Universities, Nanning 530021, Guangxi, China.,Institute of Urology and Nephrology, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, China
| |
Collapse
|
158
|
Salinas J, O’Donnell A, Kojis DJ, Pase MP, DeCarli C, Rentz DM, Berkman LF, Beiser A, Seshadri S. Association of Social Support With Brain Volume and Cognition. JAMA Netw Open 2021; 4:e2121122. [PMID: 34398201 PMCID: PMC8369356 DOI: 10.1001/jamanetworkopen.2021.21122] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
IMPORTANCE Cognitive resilience refers to the general capacity of cognitive processes to be less susceptible to differences in brain structure from age- and disease-related changes. Studies suggest that supportive social networks reduce Alzheimer disease and related disorder (ADRD) risk by enhancing cognitive resilience, but data on specific social support mechanisms are sparse. OBJECTIVE To examine the association of individual forms of social support with a global neuroanatomical measure of early ADRD vulnerability and cognition. DESIGN, SETTING, AND PARTICIPANTS This retrospective cross-sectional analysis used prospectively collected data from Framingham Study participants without dementia, stroke, or other neurological conditions who underwent brain magnetic resonance imaging and neuropsychological testing at the same visit. Data from this large, population-based, longitudinal cohort were collected from June 6, 1997, to December 13, 1999 (original cohort), and from September 11, 1998, to October 26, 2001 (offspring cohort). Data were analyzed from May 22, 2017, to June 1, 2021. EXPOSURES Total cerebral volume and, as a modifying exposure variable, self-reported availability of 5 types of social support measured by the Berkman-Syme Social Network Index. MAIN OUTCOMES AND MEASURES The primary outcome was a global measure of cognitive function. Cognitive resilience was defined as the modification of total cerebral volume's association with cognition, such that smaller β estimates (presented in SD units) indicate greater cognitive resilience (ie, better cognitive performance than estimated by lower total cerebral volume). RESULTS The study included 2171 adults (164 in the original cohort and 2007 in the offspring cohort; mean [SD] age, 63 [10] years; 1183 [54%] female). High listener availability was associated with greater cognitive resilience (β = 0.08, P < .001) compared with low listener availability (β = 0.20, P = .002). Overall findings persisted after adjustment for potential confounders. Other forms of social support were not significant modifiers (advice: β = -0.04; P = .40 for interaction; love-affection: β = -0.07, P = .28 for interaction; emotional support: β = -0.02, P = .73 for interaction; and sufficient contact: β = -0.08; P = .11 for interaction). CONCLUSIONS AND RELEVANCE The results of this cross-sectional cohort study suggest that social support in the form of supportive listening is associated with greater cognitive resilience, independently modifying the association between lower total cerebral volume and poorer cognitive function that would otherwise indicate increased ADRD vulnerability at the preclinical stage. A refined understanding of social support mechanisms has the potential to inform strategies to reduce ADRD risk and enhance cognitive resilience.
Collapse
Affiliation(s)
- Joel Salinas
- Center for Cognitive Neurology, Department of Neurology, New York University Grossman School of Medicine, New York
- The Framingham Study, Boston, Massachusetts
- Harvard Center for Population and Development Studies, Harvard University, Cambridge, Massachusetts
| | - Adrienne O’Donnell
- The Framingham Study, Boston, Massachusetts
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Daniel J. Kojis
- The Framingham Study, Boston, Massachusetts
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Matthew P. Pase
- The Framingham Study, Boston, Massachusetts
- Harvard T. H. Chan School of Public Health, Boston, Massachusetts
- Turner Institute for Brain and Mental Health, Monash University, Clayton, Victoria, Australia
| | | | - Dorene M. Rentz
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Lisa F. Berkman
- Harvard Center for Population and Development Studies, Harvard University, Cambridge, Massachusetts
- Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Alexa Beiser
- The Framingham Study, Boston, Massachusetts
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
| | - Sudha Seshadri
- The Framingham Study, Boston, Massachusetts
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio
| |
Collapse
|
159
|
Gong J, Harris K, Hackett M, Peters SAE, Brodaty H, Cooper M, Hamet P, Harrap S, Mancia G, MacMahon S, Chalmers J, Woodward M. Sex differences in risk factors for cognitive decline and dementia, including death as a competing risk, in individuals with diabetes: Results from the ADVANCE trial. Diabetes Obes Metab 2021; 23:1775-1785. [PMID: 33783955 DOI: 10.1111/dom.14391] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 03/16/2021] [Accepted: 03/25/2021] [Indexed: 12/18/2022]
Abstract
AIM To estimate the associations between risk factors and cognitive decline (CD)/dementia, and the sex differences in these risk factors in individuals with type 2 diabetes, while accounting for the competing risk of death. MATERIALS AND METHODS The Action in Diabetes and Vascular Disease: Preterax and Diamicron Modified Release Controlled Evaluation (ADVANCE) trial of 11,140 individuals with type 2 diabetes was used to estimate the odds of CD/dementia using multinomial logistic regression. RESULTS During a median 5-year follow-up, 1827 participants (43.2% women) had CD/dementia (1718 with CD only; 21 with dementia only; 88 with CD and dementia), and 929 (31.0% women) died without CD/dementia. Women had lower odds of CD/dementia than men (odds ratio [OR] [95% confidence interval], 0.88 [0.77, 1.00]); older age, higher total cholesterol, HbA1c, waist circumference, waist-to-height ratio, moderately increased albumin-creatinine ratio, stroke/transient ischaemic attack and retinal disease were each associated with greater odds of CD/dementia; higher years at education completion, baseline cognitive function, taller stature and current alcohol use were inversely associated. Higher waist circumference (women-to-men ratio of ORs [ROR], 1.05 [1.00, 1.10] per 5 cm) and presence of anxiety/depression (ROR, 1.28 [1.01, 1.63]) were associated with greater ORs for CD/dementia in women than men. CONCLUSIONS Several risk factors were associated with CD/dementia. Higher waist circumference and mental health symptoms were more strongly associated with CD/dementia in women than men. Further studies should examine the mechanisms that underlie these sex differences.
Collapse
Affiliation(s)
- Jessica Gong
- The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Katie Harris
- The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Maree Hackett
- The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia
- Faculty of Health and Wellbeing, the University of Central Lancashire, Lancashire, UK
| | - Sanne A E Peters
- The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia
- The George Institute for Global Health, Imperial College London, London, UK
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Henry Brodaty
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, New South Wales, Australia
- Dementia Centre for Research Collaboration, University of New South Wales, Sydney, New South Wales, Australia
| | - Mark Cooper
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Pavel Hamet
- Montréal Diabetes Research Centre, Centre Hospitalier de l'Université de Montréal, Quebec, Montreal, Canada
| | - Stephen Harrap
- Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | - Giuseppe Mancia
- Policlinico di Monza and IRCCS Istituto Auxologico Italiano, University of Milano-Bicocca, Milan, Italy
| | - Stephen MacMahon
- The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia
- The George Institute for Global Health, Imperial College London, London, UK
| | - John Chalmers
- The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Mark Woodward
- The George Institute for Global Health, University of New South Wales, Sydney, New South Wales, Australia
- The George Institute for Global Health, Imperial College London, London, UK
- Department of Epidemiology, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
160
|
Niu J, Iqbal K, Liu F, Hu W. Rats Display Sexual Dimorphism in Phosphorylation of Brain Tau with Age. J Alzheimers Dis 2021; 82:855-869. [PMID: 34092647 DOI: 10.3233/jad-210341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Women have a two-fold higher risk than men to Alzheimer's disease (AD) at midlife. Larger brain tau burden was consistently shown in older women than age-matched men. The biological basis for this gender disparity remains elusive. OBJECTIVE We sought to know whether tau expression and phosphorylation physiologically differ between males and females. METHODS We used western blots and immunohistochemistry to compare the levels of total tau and phosphorylated tau in the hippocampus and entorhinal cortex (EC) between sexes in Wistar rats at 40 days, and 8 and 20 months of age. RESULTS We detected no statistically significant difference in total tau, 3R-tau, and 4R-tau between sexes. However, female rats exhibited lower levels of tau unphosphorylated at the Tau-1 site at 40 days of age. At 8 months of age, females showed higher levels of tau phosphorylated at Ser190, Ser387, and Ser395 (Ser199, Ser396, and Ser404 of human tau, respectively) than males in EC. At 20 months of age, both brain regions of female rats consistently showed higher levels than males of tau phosphorylated at Ser253, Ser387, PHF-1 (Ser387/395), and Ser413 sites, which correspond to Ser262, Ser396, Ser396/404, and Ser422 of human tau, respectively. CONCLUSION Rats of both sexes have comparable levels of total tau, 3R-tau, and 4R-tau, whereas females exhibit higher levels of tau phosphorylated at multiple sites that are implicated in AD tau pathology, indicating a sexual dimorphism of tau phosphorylation that may potentially underlie the disparity in brain tau burden and risk for AD between sexes.
Collapse
Affiliation(s)
- Jiahui Niu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Khalid Iqbal
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Wen Hu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| |
Collapse
|
161
|
Sindi S, Kåreholt I, Ngandu T, Rosenberg A, Kulmala J, Johansson L, Wetterberg H, Skoog J, Sjöberg L, Wang H, Fratiglioni L, Skoog I, Kivipelto M. Sex differences in dementia and response to a lifestyle intervention: Evidence from Nordic population-based studies and a prevention trial. Alzheimers Dement 2021; 17:1166-1178. [PMID: 34255432 PMCID: PMC8361986 DOI: 10.1002/alz.12279] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Evidence on sex differences in the risk for dementia has been mixed. The goal was to assess sex differences in the development of dementia, and in the effects of a lifestyle intervention. METHODS Two strategies were adopted, one using combined data from three large Nordic population-based cohort studies (n = 2289), adopting dementia as outcome, and 2-year multidomain lifestyle intervention (n = 1260), adopting cognitive change as outcome. RESULTS There was higher risk for dementia after age 80 years in women. The positive effects of the lifestyle intervention on cognition did not significantly differ between men and women. Sex-specific analyses suggested that different vascular, lifestyle, and psychosocial risk factors are important for women and men in mid- and late-life. CONCLUSION Women had higher risk for dementia among the oldest individuals. Lifestyle interventions may be effectively implemented among older men and women.
Collapse
Affiliation(s)
- Shireen Sindi
- Division of Clinical GeriatricsCenter for Alzheimer ResearchKarolinska Institutet and Karolinska University HospitalStockholmSweden
- Ageing Epidemiology (AGE) Research UnitSchool of Public HealthImperial College LondonLondonUK
| | - Ingemar Kåreholt
- Aging Research CenterKarolinska Institutet and Stockholm UniversityStockholmSweden
- Institute of GerontologySchool of Health and WelfareAging Research Network – Jönköping (ARN‐J)Jönköping UniversityJönköpingSweden
| | - Tiia Ngandu
- Public Health Promotion UnitNational Institute for Health and WelfareHelsinkiFinland
| | - Anna Rosenberg
- Institute of Public Health and Clinical Nutrition and Institute of Clinical MedicineNeurologyUniversity of Eastern FinlandKuopioFinland
| | - Jenni Kulmala
- Division of Clinical GeriatricsCenter for Alzheimer ResearchKarolinska Institutet and Karolinska University HospitalStockholmSweden
- Public Health Promotion UnitNational Institute for Health and WelfareHelsinkiFinland
| | - Lena Johansson
- Institute of Neuroscience and PhysiologyCenter for Health and Ageing AGECAPSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Hanna Wetterberg
- Institute of Neuroscience and PhysiologyCenter for Health and Ageing AGECAPSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Johan Skoog
- Department of PsychologyCenter for Health and Ageing (AGECAP)University of GothenburgGothenburgSweden
| | - Linnea Sjöberg
- Aging Research CenterKarolinska Institutet and Stockholm UniversityStockholmSweden
| | - Hui‐Xin Wang
- Aging Research CenterKarolinska Institutet and Stockholm UniversityStockholmSweden
- Stress Research InstituteStockholom UniversityStockholmSweden
| | - Laura Fratiglioni
- Aging Research CenterKarolinska Institutet and Stockholm UniversityStockholmSweden
- Stockholm Gerontology Research CenterStockholmSweden
| | - Ingmar Skoog
- Institute of Neuroscience and PhysiologyCenter for Health and Ageing AGECAPSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Miia Kivipelto
- Division of Clinical GeriatricsCenter for Alzheimer ResearchKarolinska Institutet and Karolinska University HospitalStockholmSweden
- Ageing Epidemiology (AGE) Research UnitSchool of Public HealthImperial College LondonLondonUK
- Institute of Public Health and Clinical Nutrition and Institute of Clinical MedicineNeurologyUniversity of Eastern FinlandKuopioFinland
- Theme AgingKarolinska University HospitalStockholmSweden
| |
Collapse
|
162
|
Suanrueang P, Shen YJ, Lin HF, Er TK, Suen MW, Shieh FA. Gender differences in geriatric syndromes as mental illness and nervous system diseases in hospitalized Thai older patients. Psychogeriatrics 2021; 21:453-465. [PMID: 33847418 DOI: 10.1111/psyg.12679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 01/23/2021] [Accepted: 02/14/2021] [Indexed: 01/29/2023]
Abstract
BACKGROUND Older persons are affected by mental and neurological disorders differently, and gender plays a significant role influencing geriatric disorder differentiation. Accordingly this study characterized gender differences in geriatric syndromes among hospitalized elderly Thai patients. METHODS Probabilities of disease occurrence reflecting gender differences were calculated using historical data obtained from the Ministry of Public Health website, Thailand. We selected older patients aged 60 years and above admitted to inpatient departments in public hospitals with mental disorders and nervous system diseases from 2014 to 2018, counting over 160 000 cases each year. Descriptive statistics and odds ratios (ORs) were used to analyse and demonstrate gender differences. RESULTS Compared to older females, older males had higher occurrences of four mental disorders revealed by OR and 95% confidence interval (CI) values: substance abuse (5.74, 5.08-6.49), alcohol use (5.66, 5.44-5.89), behavioural problems (1.34, 1.31-1.37), and schizophrenia (1.10, 1.06-1.14). Lower incidences for older males were seen in the same values for three mental disorders: neurotic issues (0.46, 0.44-0.49), mood disorders (0.58, 0.56-0.60), and dementia (0.91, 0.88-0.94). For neurological disorders, men had similar higher incidences for epilepsy (1.67, 1.63-1.72), cerebral palsy (1.61, 1.57-1.65), nervous system inflammatory diseases (1.53, 1.46-1.60), ischaemic attacks (1.42, 1.36-1.48), miscellaneous other nervous disorders (1.20; 1.18-1.22), and Parkinson's disease (1.15, 1.12-1.19). By contrast, older men had lower incidences of multiple sclerosis (0.55, 0.35-0.86), migraines (0.66, 0.62-0.70), and Alzheimer's disease (0.75, 0.71-0.78). CONCLUSION Accurate characterization of gender differences in geriatric syndromes can better inform policies for appropriate early detection and prevention, and contribute to the development of treatment and intervention for various issues affecting elderly men and women's health.
Collapse
Affiliation(s)
- Passakorn Suanrueang
- Department of Healthcare Administration Specialty in Psychology, College of Medical and Health Science, Asia University, Taichung City, Taiwan
| | - Yong-Jiang Shen
- Professor of School of Educational Science, Nantong University, Nantong, China
| | - Hsiao-Fang Lin
- Associate Professor of Department and Graduate Institute of Early Childhood Development and Education, Chaoyang University of Technology, Taichung City, Taiwan
| | - Tze-Kiong Er
- Associate Professor of Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung City, Taiwan.,Division of Laboratory Medicine, Asia University Hospital, Asia University, Taichung City, Taiwan
| | - Mein-Woei Suen
- Department of Psychology, College of Medical and Health Science, Asia University, Taichung City, Taiwan.,Gender Equality Education and Research Center, Asia University, Taichung City, Taiwan.,Department of Medical Research, Asia University Hospital, Asia University, Taichung City, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung City, Taiwan
| | - Fu-An Shieh
- Lecturer of Department of Food and Beverage Management, Jin-Wen University of Science and Technology, New Taipei City, Taiwan
| |
Collapse
|
163
|
Palta P, Rippon B, Tahmi M, Pardo M, Johnson A, Tomljanovic Z, He H, Laing KK, Razlighi QR, Teresi JA, Moreno H, Brickman AM, Kreisl WC, Luchsinger JA. Sex differences in in vivo tau neuropathology in a multiethnic sample of late middle-aged adults. Neurobiol Aging 2021; 103:109-116. [PMID: 33894641 PMCID: PMC8178209 DOI: 10.1016/j.neurobiolaging.2021.03.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 03/03/2021] [Accepted: 03/13/2021] [Indexed: 10/24/2022]
Abstract
It is unclear whether women have higher brain tau pathology. The objective of this study was to examine whether women have higher tau burden than men, and whether tau differences are independent of amyloid β (Aβ) burden. We conducted a cross-sectional analysis of a multiethnic sample of 252 nondemented late middle-aged (mean age: 64.1 years) adults with tau and amyloid Positron Emission Tomography (PET) data. Tau burden was measured as global standardized uptake value ratio (SUVR) in the middle/inferior temporal gyri and medial temporal cortex with 18F-MK-6240 PET. Aβ was measured as global SUVR with 18F-Florbetaben PET. Women had higher middle/inferior temporal gyri tau SUVR compared to men. However, no sex differences in the medial temporal cortex were observed. Women had higher brain Aβ SUVR compared to men. Continuous Aβ SUVR was positively correlated with medial temporal cortex and middle/inferior temporal gyri tau SUVR. However, there was no evidence of effect modification by Aβ SUVR on sex and tau. Compared with men, women in late middle age show higher tau burden, independent of Aβ.
Collapse
Affiliation(s)
- Priya Palta
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA; Department of Epidemiology, Joseph P. Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY, USA.
| | - Brady Rippon
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Mouna Tahmi
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Michelle Pardo
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Aubrey Johnson
- Department of Neurology, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Zeljko Tomljanovic
- Department of Neurology, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Hengda He
- Department of Neurology, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Krystal K Laing
- Department of Neurology, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA; Department of Neurology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Qolamreza R Razlighi
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA; Department of Neurology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Jeanne A Teresi
- Columbia University Stroud Center at New York State Psychiatric Institute, New York, NY and Research Division, Hebrew Home in Riverdale, Bronx, NY, USA
| | - Herman Moreno
- Columbia University Stroud Center at New York State Psychiatric Institute, New York, NY and Research Division, Hebrew Home in Riverdale, Bronx, NY, USA
| | - Adam M Brickman
- Department of Neurology, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA; Gertrude H. Sergievsky Center, Columbia University Irving Medical Center, New York, NY, USA
| | - William C Kreisl
- Department of Neurology, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA; Gertrude H. Sergievsky Center, Columbia University Irving Medical Center, New York, NY, USA; Department of Neurology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - José A Luchsinger
- Department of Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA; Department of Epidemiology, Joseph P. Mailman School of Public Health, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
164
|
Schneider AC, Moon C, Whitaker K, Zhang D, Carr LJ, Bao W, Xiao Q. Association of Sleep With Risk of Alzheimer's Disease Mortality: NIH-AARP Diet and Health Study. J Appl Gerontol 2021; 41:1057-1065. [PMID: 34109847 DOI: 10.1177/07334648211019207] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVES Alzheimer's disease (AD) and related dementias contribute to one in three senior deaths. Lifestyle factors, including sleep, may contribute to AD risk and mortality; however, current evidence on sleep and AD mortality is mixed. METHODS We used data from the NIH-AARP Diet and Health Study. Sleep duration and napping were self-reported and AD death were ascertained via linkage to the National Death Index. RESULTS Long sleep and napping were both associated with increased AD mortality. Specifically, 9+ hr of sleep was associated with 50% increase (hazard ratio = 1.50, 95% CI = [1.17, 1.92]) in AD mortality when compared 7 to 8 hr, while napping for 1+ hr was associated with 29% increase (1.29 [1.08, 1.55]) when compared with no napping. Results appeared to be stronger in men and remained after removing AD deaths within first 5 years after baseline. DISCUSSION Long sleep and napping may predict higher AD mortality in the older population.
Collapse
Affiliation(s)
| | | | | | | | | | - Wei Bao
- University of Iowa, Iowa City, USA
| | - Qian Xiao
- University of Iowa, Iowa City, USA
- The University of Texas Health Science Center at Houston, USA
| |
Collapse
|
165
|
Horst BR, Furlano JA, Wong MYS, Ford SD, Han BB, Nagamatsu LS. Identification of Demographic Variables Influencing Dementia Literacy and Risk Perception Through a Global Survey. Front Public Health 2021; 9:660600. [PMID: 34169056 PMCID: PMC8217864 DOI: 10.3389/fpubh.2021.660600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/11/2021] [Indexed: 11/13/2022] Open
Abstract
Dementia literacy is important for risk mitigation and preventative strategies before disease onset. The aim of our study was to investigate dementia literacy and how demographic characteristics influence these perceptions in order to provide evidence for how dementia-centered public health initiatives should structure their focus. We conducted a globally administered online survey, through Amazon Mechanical Turk (mTurk). Survey items evaluated: (1) personal perception on the preventability of dementia, and (2) risk awareness of lifestyle factors. Differences in risk scoring between the 598 respondents were compared using Kruskal-Wallis testing factored by demographic categorizations. Most of the sample demonstrated understanding that lifestyle factors contribute some risk toward dementia, though these risk scores were generally low. Differences in risk scoring varied by demographic characteristics. Women, older adults, those with non-post-secondary attainment, below average income, and White background tended to report lower risk scores. Public health education and initiatives for dementia prevention should focus on lifestyle risk factors, in addition to considering the barriers related to the demographic factors identified that may prevent populations from accessing programs and information.
Collapse
Affiliation(s)
- Becky R Horst
- Exercise Mobility and Brain Health Lab, Department of Neuroscience, Western University, London, ON, Canada
| | - Joyla A Furlano
- Exercise Mobility and Brain Health Lab, Department of Neuroscience, Western University, London, ON, Canada
| | - Michelle Y S Wong
- Exercise Mobility and Brain Health Lab, School of Kinesiology, Western University, London, ON, Canada
| | - Sabrina D Ford
- Exercise Mobility and Brain Health Lab, Department of Neuroscience, Western University, London, ON, Canada
| | - Brenna B Han
- Exercise Mobility and Brain Health Lab, School of Kinesiology, Western University, London, ON, Canada
| | - Lindsay S Nagamatsu
- Exercise Mobility and Brain Health Lab, School of Kinesiology, Western University, London, ON, Canada
| |
Collapse
|
166
|
Androvičová R, Pfaus JG, Ovsepian SV. Estrogen pendulum in schizophrenia and Alzheimer's disease: Review of therapeutic benefits and outstanding questions. Neurosci Lett 2021; 759:136038. [PMID: 34116197 DOI: 10.1016/j.neulet.2021.136038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/21/2021] [Accepted: 06/06/2021] [Indexed: 12/29/2022]
Abstract
Although produced largely in the periphery, gonadal steroids play a key role in regulating the development and functions of the central nervous system and have been implicated in several chronic neuropsychiatric disorders, with schizophrenia and Alzheimer's disease (AD) most prominent. Despite major differences in pathobiology and clinical manifestations, in both conditions, estrogen transpires primarily with protective effects, buffering the onset and progression of diseases at various levels. As a result, estrogen replacement therapy (ERT) emerges as one of the most widely discussed adjuvant interventions. In this review, we revisit evidence supporting the protective role of estrogen in schizophrenia and AD and consider putative cellular and molecular mechanisms. We explore the underlying functional processes relevant to the manifestation of these devastating conditions, with a focus on synaptic transmission and plasticity mechanisms. We discuss specific effects of estrogen deficit on neurotransmitter systems such as cholinergic, dopaminergic, serotoninergic, and glutamatergic. While the evidence from both, preclinical and clinical reports, in general, are supportive of the protective effects of estrogen from cognitive decline to synaptic pathology, numerous questions remain, calling for further research.
Collapse
Affiliation(s)
- Renáta Androvičová
- Department of Applied Neuroscience and Neuroimaging (RA) and Department of Experimental Neuroscience (SVO), National Institute of Mental Health, Topolová 748, 250 67 Klecany, Czech Republic.
| | - James G Pfaus
- Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa, Mexico
| | - Saak V Ovsepian
- Department of Applied Neuroscience and Neuroimaging (RA) and Department of Experimental Neuroscience (SVO), National Institute of Mental Health, Topolová 748, 250 67 Klecany, Czech Republic
| |
Collapse
|
167
|
Hispanic ethnicity and in-hospital morbidity and mortality outcomes in Alzheimer's Disease: A U.S. National Study 2005-2015. Clin Neurol Neurosurg 2021; 207:106753. [PMID: 34126451 DOI: 10.1016/j.clineuro.2021.106753] [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: 12/05/2020] [Revised: 04/17/2021] [Accepted: 06/04/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND Hispanics are one of the largest and fastest-growing population in the United States. Having been reported as one of the high-risk ethnicities to develop Alzheimer's Disease (AD) makes elder Hispanics one of the significant groups of AD in the country, indicating a need to study the disparities in Hispanics vs. non-Hispanics patients. We aimed to determine the prevalence, morbidity, and mortality outcomes of AD in Hispanics. METHODS We surveyed Healthcare Cost and Utilization Project (HCUP) from 2005 to 2015 to identify patients older than 50 years who were admitted for any reason and had AD diagnosis. Prevalence, demographics, age brackets, in-hospital deaths, disease severity, and hospital length of stay (LOS) were compared between the Hispanics and Non-Hispanics. RESULTS Among 14,135,560 Hispanic discharges, 2.76% had AD, compared with 207,515,260 discharges in Non-Hispanic with 2.61% AD, p < 0.001. Hispanics had significantly more AD in all age brackets, especially over 90 years of age, p < 0.001. A significantly higher prevalence of AD in both Hispanic Females (3.27% vs. 3.10%) and Males (2.17% vs. 2.04%) was noticed, p < 0.001. In northeast and south regions of the country and urban hospitals, AD was more among Hispanics (p < 0.001). Hispanic patients were younger (81.8 ± 7.77 vs. 82.6 ± 7.50, p < 0.001), had longer LOS (6.41 ± 7.72 vs. 6.08 ± 7.05, p < 0.001), had higher hospital charges ($45,989 vs. $37,688, p < 0.001). Hispanic AD patients had higher disease severity and mortality risk (p < 0.001). However, the inpatient mortality was not different between the Hispanic and non-Hispanics. Multivariate analysis showed that Hispanics had the highest AD prevalence in the inpatient setting (OR, 1.38; 95% CI, 1.37-1.39, p < 0.001). CONCLUSION The prevalence of AD was significantly higher in inpatient Hispanics than non-Hispanics. Hispanic AD patients had a younger age compared with non-Hispanic AD. Disease severity and mortality risks were higher in Hispanics with AD than non-Hispanics with AD. However, no difference was seen in mortality rate during admission in Hispanics vs. non-Hispanics.
Collapse
|
168
|
Zhu D, Montagne A, Zhao Z. Alzheimer's pathogenic mechanisms and underlying sex difference. Cell Mol Life Sci 2021; 78:4907-4920. [PMID: 33844047 PMCID: PMC8720296 DOI: 10.1007/s00018-021-03830-w] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/09/2021] [Accepted: 03/29/2021] [Indexed: 01/01/2023]
Abstract
AD is a neurodegenerative disease, and its frequency is often reported to be higher for women than men: almost two-thirds of patients with AD are women. One prevailing view is that women live longer than men on average of 4.5 years, plus there are more women aged 85 years or older than men in most global subpopulations; and older age is the greatest risk factor for AD. However, the differences in the actual risk of developing AD for men and women of the same age is difficult to assess, and the findings have been mixed. An increasing body of evidence from preclinical and clinical studies as well as the complications in estimating incidence support the sex-specific biological mechanisms in diverging AD risk as an important adjunct explanation to the epidemiologic perspective. Although some of the sex differences in AD prevalence are due to differences in longevity, other distinct biological mechanisms increase the risk and progression of AD in women. These risk factors include (1) deviations in brain structure and biomarkers, (2) psychosocial stress responses, (3) pregnancy, menopause, and sex hormones, (4) genetic background (i.e., APOE), (5) inflammation, gliosis, and immune module (i.e., TREM2), and (6) vascular disorders. More studies focusing on the underlying biological mechanisms for this phenomenon are needed to better understand AD. This review presents the most recent data in sex differences in AD-the gateway to precision medicine, therefore, shaping expert perspectives, inspiring researchers to go in new directions, and driving development of future diagnostic tools and treatments for AD in a more customized way.
Collapse
Affiliation(s)
- Donghui Zhu
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA.
- Neuroscience Graduate Program, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA.
| | - Axel Montagne
- UK Dementia Research Institute, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Zhen Zhao
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
169
|
Wu X, Fan L, Ke S, He Y, Zhang K, Yang S. Longitudinal Associations of Stroke With Cognitive Impairment Among Older Adults in the United States: A Population-Based Study. Front Public Health 2021; 9:637042. [PMID: 34095050 PMCID: PMC8170040 DOI: 10.3389/fpubh.2021.637042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 04/06/2021] [Indexed: 11/24/2022] Open
Abstract
Objective: The aim of this study was to explore the longitudinal associations of stroke with cognitive impairment in older US adults. Method: The data used in this longitudinal analysis were extracted from the National Health and Aging Trends Study (NHATS) from 2011 to 2019. Univariate and multivariable Cox proportional hazards regression models were used to estimate the longitudinal association of stroke with cognitive impairment. The multivariable model was adjusted by demographic, physical, and mental characteristics, and the complex survey design of NHATS was taken into consideration. Results: A total of 7,052 participants with complete data were included. At the baseline, the weighted proportion of cognitive impairment was 19.37% (95% CI, 17.92–20.81%), and the weighted proportion of the history of stroke was 9.81% (95% CI, 8.90–10.72%). In univariate analysis, baseline stroke history was significantly associated with cognitive impairment in the future (hazard ratio, 1.746; 95% CI, 1.461–2.088), and the baseline cognitive impairment was significantly associated with future report of stroke (hazard ratio, 1.436; 95% CI, 1.088–1.896). In multivariable model, stroke was also significantly associated with cognitive impairment (hazard ratio, 1.241; 95% CI, 1.011–1.522); however, the reverse association was not significant (hazard ratio, 1.068; 95% CI, 0.788–1.447). After the data from proxy respondents were excluded, in the sensitive analyses, the results remained unchanged. Conclusion: Older adults in the United States who suffered strokes are more likely to develop cognitive impairment as a result in the future than those who have not had strokes. However, the reverse association did not hold. Furthermore, the study suggests that it is necessary to screen and take early intervention for cognitive impairment in stroke survivors and prevent the incidence of stroke by modifying risk factors in the general population with rapidly growing older US adults.
Collapse
Affiliation(s)
- Xia Wu
- Department of Otorhinolaryngology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Li Fan
- Department of Orthopaedics, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Songqing Ke
- Ministry of Education Key Laboratory of Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yangting He
- Ministry of Education Key Laboratory of Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ke Zhang
- Biostatistician at Causality Clinical Data Technology Co., Ltd, Wuhan, China
| | - Shijun Yang
- Department of Cardiology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
170
|
Multimodal neuroimaging of sex differences in cognitively impaired patients on the Alzheimer's continuum: greater tau-PET retention in females. Neurobiol Aging 2021; 105:86-98. [PMID: 34049062 DOI: 10.1016/j.neurobiolaging.2021.04.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 04/03/2021] [Accepted: 04/05/2021] [Indexed: 12/23/2022]
Abstract
We assessed sex differences in amyloid- and tau-PET retention in 119 amyloid positive patients with mild cognitive impairment or Alzheimer's disease (AD) dementia. Patients underwent 3T-MRI, 11C-PIB amyloid-PET and 18F-Flortaucipir tau-PET. Linear ordinary least squares regression models tested sex differences in Flortaucipir-PET SUVR in a summary temporal region of interest as well as global PIB-PET. No sex differences were observed in demographics, Clinical Dementia Rating Sum of Boxes (CDR-SoB), Mini-Mental State Exam (MMSE), raw episodic memory scores, or cortical thickness. Females had higher global PIB SUVR (ηp²=.043, p=.025) and temporal Flortaucipir SUVR (ηp²=.070, p=.004), adjusting for age and CDR-SoB. Sex differences in temporal Flortaucipir-PET remained significant when controlling additionally for PIB SUVR and APOE4 status (ηp²=.055, p=.013), or when using partial volume-corrected data. No sex differences were present in areas of known Flortaucipir off-target binding. Overall, females demonstrated greater AD regional tau-PET burden than males despite clinical comparability. Further characterization of sex differences will provide insight into AD pathogenesis and support development of personalized therapeutic strategies.
Collapse
|
171
|
Ruediger SL, Koep JL, Keating SE, Pizzey FK, Coombes JS, Bailey TG. Effect of menopause on cerebral artery blood flow velocity and cerebrovascular reactivity: Systematic review and meta-analysis. Maturitas 2021; 148:24-32. [PMID: 34024348 DOI: 10.1016/j.maturitas.2021.04.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/28/2021] [Accepted: 04/11/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND Menopause and its associated decline in oestrogen is linked to chronic conditions like cardiovascular disease and osteoporosis, which may be difficult to disentangle from the effects of ageing. Further, post-menopausal women are at increased risk of cerebrovascular disease, linked to declines in cerebral blood flow (CBF) and cerebrovascular reactivity (CVR), yet the direct understanding of the impact of the menopause on cerebrovascular function is unclear. The aim of this systematic review and meta-analysis was to examine the literature investigating CBF and CVR in pre- compared with post-menopausal women METHODS: Five databases were searched for studies assessing CBF or CVR in pre- and post-menopausal women. Meta-analysis examined the effect of menopausal status on middle cerebral artery velocity (MCAv), and GRADE-assessed evidence certainty RESULTS: Nine studies (n=504) included cerebrovascular outcomes. Six studies (n=239) reported negligible differences in MCAv between pre- and post-menopausal women [2.11cm/s (95% CI: -8.94 to 4.73, p=0.54)], but with a "low" certainty of evidence. MCAv was lower in post-menopausal women in two studies, when MCAv was adjusted for blood pressure. CVR was lower in post- compared with pre-menopausal women in two of three studies, but high-quality evidence is lacking. Across outcomes, study methodology and reporting criteria for menopause were inconsistent CONCLUSIONS: MCAv was similar in post- compared with pre-menopausal women. Methodological differences in characterising menopause and inconsistent reporting of cerebrovascular outcomes make comparisons difficult. Comprehensive assessments of cerebrovascular function of the intra- and extracranial arteries to determine the physiological implications of menopause on CBF with healthy ageing is warranted.
Collapse
Affiliation(s)
- Stefanie L Ruediger
- Physiology and Ultrasound Laboratory in Science and Exercise, Centre for Research on Exercise, Physical Activity and Health; School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Jodie L Koep
- Physiology and Ultrasound Laboratory in Science and Exercise, Centre for Research on Exercise, Physical Activity and Health; School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, QLD, Australia; Children's Health and Exercise Research Centre, Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter, UK
| | - Shelley E Keating
- Physiology and Ultrasound Laboratory in Science and Exercise, Centre for Research on Exercise, Physical Activity and Health; School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Faith K Pizzey
- Physiology and Ultrasound Laboratory in Science and Exercise, Centre for Research on Exercise, Physical Activity and Health; School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Jeff S Coombes
- Physiology and Ultrasound Laboratory in Science and Exercise, Centre for Research on Exercise, Physical Activity and Health; School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Tom G Bailey
- Physiology and Ultrasound Laboratory in Science and Exercise, Centre for Research on Exercise, Physical Activity and Health; School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, QLD, Australia; School of Nursing, Midwifery and Social Work, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
172
|
Abstract
This article describes the public health impact of Alzheimer's disease (AD), including incidence and prevalence, mortality and morbidity, use and costs of care, and the overall impact on caregivers and society. The Special Report discusses the challenges of providing equitable health care for people with dementia in the United States. An estimated 6.2 million Americans age 65 and older are living with Alzheimer's dementia today. This number could grow to 13.8 million by 2060 barring the development of medical breakthroughs to prevent, slow or cure AD. Official death certificates recorded 121,499 deaths from AD in 2019, the latest year for which data are available, making Alzheimer's the sixth-leading cause of death in the United States and the fifth-leading cause of death among Americans age 65 and older. Between 2000 and 2019, deaths from stroke, heart disease and HIV decreased, whereas reported deaths from AD increased more than 145%. This trajectory of deaths from AD was likely exacerbated in 2020 by the COVID-19 pandemic. More than 11 million family members and other unpaid caregivers provided an estimated 15.3 billion hours of care to people with Alzheimer's or other dementias in 2020. These figures reflect a decline in the number of caregivers compared with a decade earlier, as well as an increase in the amount of care provided by each remaining caregiver. Unpaid dementia caregiving was valued at $256.7 billion in 2020. Its costs, however, extend to family caregivers' increased risk for emotional distress and negative mental and physical health outcomes - costs that have been aggravated by COVID-19. Average per-person Medicare payments for services to beneficiaries age 65 and older with AD or other dementias are more than three times as great as payments for beneficiaries without these conditions, and Medicaid payments are more than 23 times as great. Total payments in 2021 for health care, long-term care and hospice services for people age 65 and older with dementia are estimated to be $355 billion. Despite years of efforts to make health care more equitable in the United States, racial and ethnic disparities remain - both in terms of health disparities, which involve differences in the burden of illness, and health care disparities, which involve differences in the ability to use health care services. Blacks, Hispanics, Asian Americans and Native Americans continue to have a higher burden of illness and lower access to health care compared with Whites. Such disparities, which have become more apparent during COVID-19, extend to dementia care. Surveys commissioned by the Alzheimer's Association recently shed new light on the role of discrimination in dementia care, the varying levels of trust between racial and ethnic groups in medical research, and the differences between groups in their levels of concern about and awareness of Alzheimer's disease. These findings emphasize the need to increase racial and ethnic diversity in both the dementia care workforce and in Alzheimer's clinical trials.
Collapse
|
173
|
Kuang W, Gao M, Tian L, Wan Y, Qiu P. Trends in the prevalence of cognitive impairment in Chinese older adults: based on the Chinese Longitudinal Healthy Longevity Survey cohorts from 1998 to 2014. Int Health 2021; 12:378-387. [PMID: 31967316 PMCID: PMC7443732 DOI: 10.1093/inthealth/ihz114] [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: 12/12/2018] [Revised: 10/09/2019] [Accepted: 10/30/2019] [Indexed: 02/05/2023] Open
Abstract
In the context of a rapidly ageing Chinese population, this study aims to examine trends in the prevalence of cognitive impairment among people ≥65 y of age in China. Our sample is 72 821 adults aged 65–105 y from the seven waves of the Chinese Longitudinal Healthy Longevity Survey, a national mixed longitudinal cohort. The Chinese version of the Mini-Mental State Examination was used to measure CI. Risk factor-adjusted prevalence trend was examined using multilevel regression models. Age-standardized prevalence of cognitive impairment increased from 11.00% in 1998 to 11.84% in 2008 and decreased to 8.88% in 2014. Older age, female gender, less education, rural residence, not married, lack of physical and cognitive activities, suffering from stroke, vision and hearing impairment, and activities of daily living disability were negatively associated with cognitive impairment. Our study suggests a decreasing trend of cognitive impairment prevalence in China. However, whether decreasing prevalence will contribute to a reduced burden of cognitive impairment given the ageing of the population is unknown.
Collapse
Affiliation(s)
- Weihong Kuang
- West China Hospital, Sichuan University, 37, Guoxuexiang, Chengdu, China
| | - Mingyue Gao
- Centre for Pediatric Epidemiology and Biostatistics, Institute of Child Health, University College London, 30 Guilford St, Holborn, London WC1N 1EH, United Kingdom
| | - Liantian Tian
- West China School of Public Health/ No. 4 West China Teaching Hospital, Sichuan University, Chengdu, China
| | - Yang Wan
- West China School of Public Health/ No. 4 West China Teaching Hospital, Sichuan University, Chengdu, China
| | - Peiyuan Qiu
- West China School of Public Health/ No. 4 West China Teaching Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
174
|
Bosoi CR, Vandal M, Tournissac M, Leclerc M, Fanet H, Mitchell PL, Verreault M, Trottier J, Virgili J, Tremblay C, Lippman HR, Bajaj JS, Barbier O, Marette A, Calon F. High-Fat Diet Modulates Hepatic Amyloid β and Cerebrosterol Metabolism in the Triple Transgenic Mouse Model of Alzheimer's Disease. Hepatol Commun 2021; 5:446-460. [PMID: 33681678 PMCID: PMC7917280 DOI: 10.1002/hep4.1609] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/09/2020] [Accepted: 08/20/2020] [Indexed: 12/19/2022] Open
Abstract
Obesity and diabetes are strongly associated not only with fatty liver but also cognitive dysfunction. Moreover, their presence, particularly in midlife, is recognized as a risk factor for Alzheimer's disease (AD). AD, the most common cause of dementia, is increasingly considered as a metabolic disease, although underlying pathogenic mechanisms remain unclear. The liver plays a major role in maintaining glucose and lipid homeostasis, as well as in clearing the AD neuropathogenic factor amyloid-β (Aβ) and in metabolizing cerebrosterol, a cerebral-derived oxysterol proposed as an AD biomarker. We hypothesized that liver impairment induced by obesity contributes to AD pathogenesis. We show that the AD triple transgenic mouse model (3xTg-AD) fed a chow diet presents a hepatic phenotype similar to nontransgenic controls (NTg) at 15 months of age. A high-fat diet (HFD), started at the age of 6 months and continued for 9 months, until sacrifice, induced hepatic steatosis in NTg, but not in 3xTg-AD mice, whereas HFD did not induce changes in hepatic fatty acid oxidation, de novo lipogenesis, and gluconeogenesis. HFD-induced obesity was associated with a reduction of insulin-degrading enzyme, one of the main hepatic enzymes responsible for Aβ clearance. The hepatic rate of cerebrosterol glucuronidation was lower in obese 3xTg-AD than in nonobese controls (P < 0.05) and higher compared with obese NTg (P < 0.05), although circulating levels remained unchanged. Conclusion: Modulation of hepatic lipids, Aβ, and cerebrosterol metabolism in obese 3xTg-AD mice differs from control mice. This study sheds light on the liver-brain axis, showing that the chronic presence of NAFLD and changes in liver function affect peripheral AD features and should be considered during development of biomarkers or AD therapeutic targets.
Collapse
Affiliation(s)
- Cristina R Bosoi
- Centre De Recherche De L'institut De Cardiologie Et Pneumologie De QuébecQuébecCanada.,Faculté De MédecineUniversité LavalQuébecCanada.,Axe NeurosciencesCentre De Recherche du CHU de Québec-Université LavalQuébecCanada
| | - Milène Vandal
- Axe NeurosciencesCentre De Recherche du CHU de Québec-Université LavalQuébecCanada.,Faculté De PharmacieUniversité LavalQuébecCanada
| | - Marine Tournissac
- Axe NeurosciencesCentre De Recherche du CHU de Québec-Université LavalQuébecCanada.,Faculté De PharmacieUniversité LavalQuébecCanada.,OptiNutriBrain International Associated LaboratoryQuébecCanada
| | - Manon Leclerc
- Axe NeurosciencesCentre De Recherche du CHU de Québec-Université LavalQuébecCanada.,Faculté De PharmacieUniversité LavalQuébecCanada
| | - Hortense Fanet
- Axe NeurosciencesCentre De Recherche du CHU de Québec-Université LavalQuébecCanada.,Faculté De PharmacieUniversité LavalQuébecCanada.,OptiNutriBrain International Associated LaboratoryQuébecCanada
| | - Patricia L Mitchell
- Centre De Recherche De L'institut De Cardiologie Et Pneumologie De QuébecQuébecCanada
| | - Mélanie Verreault
- Laboratoire de Pharmacologie MoléculaireAxe Endocrinologie et NéphrologieCentre de Recherche du CHU de Québec (Pavillon CHUL)QuébecCanada
| | - Jocelyn Trottier
- Laboratoire de Pharmacologie MoléculaireAxe Endocrinologie et NéphrologieCentre de Recherche du CHU de Québec (Pavillon CHUL)QuébecCanada
| | - Jessica Virgili
- Axe NeurosciencesCentre De Recherche du CHU de Québec-Université LavalQuébecCanada.,Faculté De PharmacieUniversité LavalQuébecCanada
| | - Cynthia Tremblay
- Axe NeurosciencesCentre De Recherche du CHU de Québec-Université LavalQuébecCanada
| | - H Robert Lippman
- Central Virginia VA Health Care SystemRichmondVA.,Virginia Commonwealth UniversityRichmondVA
| | - Jasmohan S Bajaj
- Central Virginia VA Health Care SystemRichmondVA.,Virginia Commonwealth UniversityRichmondVA
| | - Olivier Barbier
- Faculté De PharmacieUniversité LavalQuébecCanada.,Laboratoire de Pharmacologie MoléculaireAxe Endocrinologie et NéphrologieCentre de Recherche du CHU de Québec (Pavillon CHUL)QuébecCanada
| | - André Marette
- Centre De Recherche De L'institut De Cardiologie Et Pneumologie De QuébecQuébecCanada.,Faculté De MédecineUniversité LavalQuébecCanada
| | - Frédéric Calon
- Axe NeurosciencesCentre De Recherche du CHU de Québec-Université LavalQuébecCanada.,Faculté De PharmacieUniversité LavalQuébecCanada.,OptiNutriBrain International Associated LaboratoryQuébecCanada
| |
Collapse
|
175
|
Smith R, Strandberg O, Mattsson-Carlgren N, Leuzy A, Palmqvist S, Pontecorvo MJ, Devous MD, Ossenkoppele R, Hansson O. The accumulation rate of tau aggregates is higher in females and younger amyloid-positive subjects. Brain 2021; 143:3805-3815. [PMID: 33439987 PMCID: PMC7805812 DOI: 10.1093/brain/awaa327] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/21/2020] [Accepted: 08/06/2020] [Indexed: 02/06/2023] Open
Abstract
The development of tau-PET allows paired helical filament tau pathology to be visualized in vivo. Increased knowledge about conditions affecting the rate of tau accumulation could guide the development of therapies halting the progression of Alzheimer’s disease. However, the factors modifying the rate of tau accumulation over time in Alzheimer’s disease are still largely unknown. Large-scale longitudinal cohort studies, adjusting for baseline tau load, are needed to establish such risk factors. In the present longitudinal study, 419 participants from four cohorts in the USA (Avid 05e, n = 157; Expedition-3, n = 82; ADNI, n = 123) and Sweden (BioFINDER, n = 57) were scanned repeatedly with tau-PET. The study participants were cognitively unimpaired (n = 153), or patients with mild cognitive impairment (n = 139) or Alzheimer’s disease dementia (n = 127). Participants underwent two to four tau-PET (18F-flortaucipir) scans with a mean (± standard deviation) of 537 (±163) days between the first and last scan. The change in tau-PET signal was estimated in temporal meta- and neocortical regions of interest. Subject specific tau-PET slopes were predicted simultaneously by age, sex, amyloid status (determined by amyloid-β PET), APOE ε4 genotype, study cohort, diagnosis and baseline tau load. We found that accelerated increase in tau-PET signal was observed in amyloid-β-positive mild cognitive impairment (3.0 ± 5.3%) and Alzheimer’s disease dementia (2.9 ± 5.7%), respectively, when compared to either amyloid-β-negative cognitively unimpaired (0.4 ± 2.7%), amyloid-β-negative mild cognitive impairment (−0.4 ± 2.3%) or amyloid-β-positive cognitively unimpaired (1.2 ± 2.8%). Tau-PET uptake was accelerated in females (temporal region of interest: t = 2.86, P = 0.005; neocortical region of interest: t = 2.90, P = 0.004), younger individuals (temporal region of interest: t = −2.49, P = 0.013), and individuals with higher baseline tau-PET signal (temporal region of interest: t = 3.83, P < 0.001; neocortical region of interest: t = 5.01, P < 0.001). Tau-PET slopes decreased with age in amyloid-β-positive subjects, but were stable by age in amyloid-β-negative subjects (age × amyloid-β status interaction: t = −2.39, P = 0.018). There were no effects of study cohort or APOE ε4 positivity. In a similar analysis on longitudinal amyloid-β-PET (in ADNI subjects only, n = 639), we found significant associations between the rate of amyloid-β accumulation and APOE ε4 positivity, older age and baseline amyloid-β positivity, but no effect of sex. In conclusion, in this longitudinal PET study comprising four cohorts, we found that the tau accumulation rate is greater in females and younger amyloid-β-positive individuals, while amyloid-β accumulation is greater in APOE ε4 carriers and older individuals. These findings are important considerations for the design of clinical trials, and might improve our understanding of factors associated with faster tau aggregation and spread.
Collapse
Affiliation(s)
- Ruben Smith
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Olof Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Antoine Leuzy
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, Lund, Sweden
| | | | | | - Rik Ossenkoppele
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Amsterdam University Medical Center, Alzheimercenter, Neuroscience Campus Amsterdam, Amsterdam, The Netherlands
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Memory Clinic, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
176
|
Tong TM, Dao TTH, Doan LP, Nguyen DT, Nguyen QTT, Do TTT, Truong KD, Phan MD, Nguyen HN, Tran TC, Giang H. Genetic analysis of Vietnamese patients with early-onset Alzheimer's disease. Int J Neurosci 2021; 132:1190-1197. [PMID: 33397166 DOI: 10.1080/00207454.2020.1870974] [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: 10/22/2022]
Abstract
Purpose of the study: Alzheimer's disease (AD) is the most common type of dementia and its prevalence is rapidly increasing worldwide. Early-onset Alzheimer's disease (EOAD) constitutes of patients with age of onset earlier than 65 year-old and is known to be associated with genetic mutations. In this study, we reported the first genetic analysis of Vietnamese patients with EOAD.Materials and methods: We analyzed targeted sequencing data obtained from a cohort of 51 Vietnamese EOAD patients to identify pathogenic variants in twenty nine well-characterized neurodengerative genes.Results: We identified four missense mutations in APP/PSEN1 genes from six individuals, which accounts for 11.8% of all tested cases. Three of these mutations were previously reported as pathogenic and one mutation in the APP gene was newly identified and might be specific for Vietnamese patients. Our study also found eight individuals carrying homozygous APOE ε4 allele, the main risk factor gene for late-onset AD.Conclusions: Our findings showed that mutation rate in APP/PSEN genes in Vietnamese EOAD patients is consistent with that in other ethnic groups. Although further functional studies are required to validate the pathogenesis of the new mutations, our study demonstrated the necessity of genetic screening for EOAD patients as well as additional genetic data collection in Vietnamese population.
Collapse
Affiliation(s)
- Trang Mai Tong
- Department of Neurology, University Medical Center, Ho Chi Minh City, Vietnam
| | | | | | | | | | | | | | - Minh-Duy Phan
- Medical Genetics Institute, Ho Chi Minh City, Vietnam
| | - Hoai-Nghia Nguyen
- Center for Molecular Biomedicine, University of Medicine and Pharmacy, Ho Chi Minh City, Vietnam
| | - Thang Cong Tran
- Department of Neurology, University of Medicine and Pharmacy, Ho Chi Minh City, Vietnam
| | - Hoa Giang
- Medical Genetics Institute, Ho Chi Minh City, Vietnam
| |
Collapse
|
177
|
Ophir K, Ran B, Felix B, Amir G. Ten year cumulative incidence of dementia after late onset epilepsy of unknown etiology. J Clin Neurosci 2021; 86:247-251. [PMID: 33775336 DOI: 10.1016/j.jocn.2021.01.030] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 12/16/2020] [Accepted: 01/18/2021] [Indexed: 11/17/2022]
Abstract
Recent epidemiological studies suggest late life onset epilepsy of unknown etiology (LOEU) is a risk factor for future dementia. These studies rely on inclusion and exclusion of multiple diagnostic codes rather than structured data and neuroimaging findings, and thus challenging to interpret clinically. We assessed the cumulative incidence of dementia in patients with LOEU diagnosed through admission data and neuroimaging over a 10-year follow-up and compared baseline characteristics that distinguish group level incident dementia. We screened our hospital records for patients aged 55-69 with new epilepsy, admitted between 2000 and 2008, and excluded patients diagnosed with epilepsy from an underlying cause on medical records or neuroimaging. We used retrospective hospital data to follow patients for incident dementia or mortality at 10 years and compared baseline (demographics, depression or anxiety, vascular risk factors, results of electroencephalogram (EEG) studies, antidepressant use and type of ant seizure medication) and follow up (seizure recurrence, incident cerebrovascular disease) characteristics for patients with and without incident dementia. Fifty-four LOEU cases were screened, age at first seizure was 61 ± 5. The 10-year cumulative incidence of dementia was 22.20% (95% Confidence Interval 22.08-23.10%) and time to dementia diagnosis was 5.4 ± 3.9 years. Patients with incident dementia were more likely to be women (83% vs 38%, p = 0.002), have interictal epileptic form discharges (IED) on baseline EEG (70% vs 29%, p = 0.011) and depression or anxiety (50% vs 18%, p = 0.026). No differences were found in other baseline or follow up characteristics. Our results support recent findings of dementia incidence in LOEU. Prospective studies on LOEU should evaluate phenotypic determinants of individuals with late life epilepsy and the rate of progression to dementia.
Collapse
Affiliation(s)
- Keret Ophir
- Global Brain Health Institute at University of California San Francisco, San Francisco, CA, USA.
| | - Brauner Ran
- Department of Neurology, Rabin Medical Center, Beilinson Campus, Petach Tikva, Israel; Cognitive Neurology Clinic, Rabin Medical Center, Beilinson Campus, Petach Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Benninger Felix
- Department of Neurology, Rabin Medical Center, Beilinson Campus, Petach Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Glik Amir
- Department of Neurology, Rabin Medical Center, Beilinson Campus, Petach Tikva, Israel; Cognitive Neurology Clinic, Rabin Medical Center, Beilinson Campus, Petach Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
178
|
Levine DA, Gross AL, Briceño EM, Tilton N, Giordani BJ, Sussman JB, Hayward RA, Burke JF, Hingtgen S, Elkind MSV, Manly JJ, Gottesman RF, Gaskin DJ, Sidney S, Sacco RL, Tom SE, Wright CB, Yaffe K, Galecki AT. Sex Differences in Cognitive Decline Among US Adults. JAMA Netw Open 2021; 4:e210169. [PMID: 33630089 PMCID: PMC7907956 DOI: 10.1001/jamanetworkopen.2021.0169] [Citation(s) in RCA: 248] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/06/2021] [Indexed: 12/11/2022] Open
Abstract
Importance Sex differences in dementia risk are unclear, but some studies have found greater risk for women. Objective To determine associations between sex and cognitive decline in order to better understand sex differences in dementia risk. Design, Setting, and Participants This cohort study used pooled analysis of individual participant data from 5 cohort studies for years 1971 to 2017: Atherosclerosis Risk in Communities Study, Coronary Artery Risk Development in Young Adults Study, Cardiovascular Health Study, Framingham Offspring Study, and Northern Manhattan Study. Linear mixed-effects models were used to estimate changes in each continuous cognitive outcome over time by sex. Data analysis was completed from March 2019 to October 2020. Exposure Sex. Main Outcomes and Measures The primary outcome was change in global cognition. Secondary outcomes were change in memory and executive function. Outcomes were standardized as t scores (mean [SD], 50 [10]); a 1-point difference represents a 0.1-SD difference in cognition. Results Among 34 349 participants, 26 088 who self-reported Black or White race, were free of stroke and dementia, and had covariate data at or before the first cognitive assessment were included for analysis. Median (interquartile range) follow-up was 7.9 (5.3-20.5) years. There were 11 775 (44.7%) men (median [interquartile range] age, 58 [51-66] years at first cognitive assessment; 2229 [18.9%] Black) and 14 313 women (median [interquartile range] age, 58 [51-67] years at first cognitive assessment; 3636 [25.4%] Black). Women had significantly higher baseline performance than men in global cognition (2.20 points higher; 95% CI, 2.04 to 2.35 points; P < .001), executive function (2.13 points higher; 95% CI, 1.98 to 2.29 points; P < .001), and memory (1.89 points higher; 95% CI, 1.72 to 2.06 points; P < .001). Compared with men, women had significantly faster declines in global cognition (-0.07 points/y faster; 95% CI, -0.08 to -0.05 points/y; P < .001) and executive function (-0.06 points/y faster; 95% CI, -0.07 to -0.05 points/y; P < .001). Men and women had similar declines in memory (-0.004 points/y faster; 95% CI, -0.023 to 0.014; P = .61). Conclusions and Relevance The results of this cohort study suggest that women may have greater cognitive reserve but faster cognitive decline than men, which could contribute to sex differences in late-life dementia.
Collapse
Affiliation(s)
- Deborah A. Levine
- Cognitive Health Services Research Program, Department of Internal Medicine, University of Michigan, Ann Arbor
- Department of Neurology and Stroke Program, University of Michigan, Ann Arbor
- Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor
| | - Alden L. Gross
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Emily M. Briceño
- Cognitive Health Services Research Program, Department of Internal Medicine, University of Michigan, Ann Arbor
- Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor
- Department of Physical Medicine and Rehabilitation, University of Michigan, Ann Arbor
| | - Nicholas Tilton
- Cognitive Health Services Research Program, Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Bruno J. Giordani
- Department of Psychiatry, University of Michigan, Ann Arbor
- Michigan Alzheimer’s Disease Center, University of Michigan, Ann Arbor
| | - Jeremy B. Sussman
- Cognitive Health Services Research Program, Department of Internal Medicine, University of Michigan, Ann Arbor
- Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor
- VA Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - Rodney A. Hayward
- Cognitive Health Services Research Program, Department of Internal Medicine, University of Michigan, Ann Arbor
- Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor
- VA Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - James F. Burke
- Department of Neurology and Stroke Program, University of Michigan, Ann Arbor
- Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor
- VA Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - Stephanie Hingtgen
- Cognitive Health Services Research Program, Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Mitchell S. V. Elkind
- Vagelos College of Physicians and Surgeons, Department of Neurology, Columbia University, New York, New York
- Mailman School of Public Health, Department of Epidemiology, Columbia University, New York, New York
| | - Jennifer J. Manly
- Vagelos College of Physicians and Surgeons, Department of Neurology, Columbia University, New York, New York
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York
| | | | - Darrell J. Gaskin
- Department of Health Policy and Management, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Stephen Sidney
- Kaiser Permanente Northern California Division of Research, Oakland
| | - Ralph L. Sacco
- Department of Neurology, University of Miami, Miami, Florida
| | - Sarah E. Tom
- Vagelos College of Physicians and Surgeons, Department of Neurology, Columbia University, New York, New York
- Mailman School of Public Health, Department of Epidemiology, Columbia University, New York, New York
| | - Clinton B. Wright
- Division of Clinical Research, National Institute of Neurological Disorders and Stroke (NINDS), Bethesda, Maryland
| | - Kristine Yaffe
- Department of Psychiatry, University of California, San Francisco
- Department of Neurology, University of California, San Francisco
- Department of Epidemiology, University of California, San Francisco
| | - Andrzej T. Galecki
- Cognitive Health Services Research Program, Department of Internal Medicine, University of Michigan, Ann Arbor
- Department of Biostatistics, University of Michigan, Ann Arbor
| |
Collapse
|
179
|
Baroni C, Lionetti V. The impact of sex and gender on heart-brain axis dysfunction: current concepts and novel perspectives. Can J Physiol Pharmacol 2021; 99:151-160. [PMID: 33002366 DOI: 10.1139/cjpp-2020-0391] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The heart-brain axis (HBA) recapitulates all the circuits that regulate bidirectional flow of communication between heart and brain. Several mechanisms may underlie the interdependent relationship involving heterogeneous tissues at rest and during specific target organ injury such as myocardial infarction, heart failure, arrhythmia, stroke, mood disorders, or dementia. In-depth translational studies of the HBA dysfunction under single-organ injury should include both male and female animals to develop sex- and gender-oriented prevention, diagnosis, and treatment strategies. Indeed, sex and gender are determining factors as females and males exhibit significant differences in terms of susceptibility to risk factors, age of onset, severity of symptoms, and outcome. Despite most studies having focused on the male population, we have conducted a careful appraisal of the literature investigating HBA in females. In particular, we have (i) analyzed sex-related heart and brain illnesses, (ii) recapitulated the most significant studies simultaneously conducted on cardio- and cerebro-vascular systems in female populations, and (iii) hypothesized future perspectives for the development of a gender-based approach to HBA dysfunction. Although sex- and gender-oriented research is at its infancy, the impact of sex on HBA dysfunction is opening unexpected new avenues for managing the health of female subjects exposed to risk of lifestyle multi-organ disease.
Collapse
Affiliation(s)
- Carlotta Baroni
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Vincenzo Lionetti
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
- UOS Anesthesiology and Intensive Care Medicine, Fondazione Toscana G. Monasterio, Pisa, Italy
| |
Collapse
|
180
|
Than S, Moran C, Beare R, Vincent AJ, Collyer TA, Wang W, Callisaya ML, Thomson R, Phan TG, Fornito A, Srikanth VK. Interactions Between Age, Sex, Menopause, and Brain Structure at Midlife: A UK Biobank Study. J Clin Endocrinol Metab 2021; 106:410-420. [PMID: 33205159 DOI: 10.1210/clinem/dgaa847] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Indexed: 11/19/2022]
Abstract
OBJECTIVES Age and female sex are risk factors for dementia, and menopause is associated with cognitive dysfunction. Previous work largely considered the effects of sex and menopause as being independent of age. We studied whether age interacts with sex or menopause in explaining imaging biomarkers of dementia during midlife. METHODS In this cross-sectional study of UK Biobank participants with brain magnetic resonance imaging (MRI), we explored the interaction of age with sex or menopausal status in explaining total brain volume (TBV), gray matter volume (GMV), white matter volume (WMV), white matter hyperintensity volume (WMHV), regional cortical volume , and subcortical volume. RESULTS Data were available for 1827 postmenopausal women, 230 pre/perimenopausal women and 2165 men (median age 63.3 years). There was a significant interaction between age and sex (P = .024) for TBV, where the inverse association age with TBV was steeper in women (β = -5.35 mL/year) than in men (β = -4.77 mL/year). Similar age-sex interactions were also observed for GMV and WMV. In women, there was a significant interaction between age and menopausal status (P = .007) where the inverse association of age with TBV was steeper in postmenopausal (β = -5.89 mL/year) than in pre/perimenopausal women (β = -1.61 mL/year). Similar age-menopause interactions were found in predicting lower GMV and higher WMHV. Differences in the direction of these age-sex and age-menopause interactions were found for regional cortical and subcortical brain volumes. CONCLUSION Sex and menopause both interact with age during midlife in explaining MRI biomarkers of dementia. Further work is required to understand the mechanisms driving these interactions to develop strategies for delaying dementia.
Collapse
Affiliation(s)
- Stephanie Than
- Academic Unit, Peninsula Clinical School, Central Clinical School, Melbourne, Monash University, Victoria, Australia
- Department of Geriatric Medicine, Peninsula Health, Melbourne, Victoria, Australia
| | - Chris Moran
- Academic Unit, Peninsula Clinical School, Central Clinical School, Melbourne, Monash University, Victoria, Australia
- Department of Geriatric Medicine, Peninsula Health, Melbourne, Victoria, Australia
| | - Richard Beare
- Academic Unit, Peninsula Clinical School, Central Clinical School, Melbourne, Monash University, Victoria, Australia
- Developmental Imaging, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Amanda J Vincent
- Monash Centre for Health Research and Implementation, School of Public Health and Preventative Medicine, Monash University, Melbourne, Victoria, Australia
- Department of Endocrinology, Monash Health, Melbourne, Victoria, Australia
| | - Taya A Collyer
- Academic Unit, Peninsula Clinical School, Central Clinical School, Melbourne, Monash University, Victoria, Australia
| | - Wei Wang
- Academic Unit, Peninsula Clinical School, Central Clinical School, Melbourne, Monash University, Victoria, Australia
- Department of Clinical Epidemiology, School of Public Health and Preventative Medicine, Cabrini Institute, Monash University, Melbourne, Victoria, Australia
| | - Michele L Callisaya
- Academic Unit, Peninsula Clinical School, Central Clinical School, Melbourne, Monash University, Victoria, Australia
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Russell Thomson
- Centre for Research in Mathematics, Western Sydney University, Sydney, New South Wales, Australia
| | - Thanh G Phan
- Stroke and Aging Research Group, School of Clinical Sciences, Monash University, Melbourne, Victoria, Australia
| | - Alex Fornito
- Monash Biomedical Imaging, School of Psychological Science, Turner Institute for Brain and Mental Health, Monash University, Melbourne, Victoria, Australia
| | - Velandai K Srikanth
- Academic Unit, Peninsula Clinical School, Central Clinical School, Melbourne, Monash University, Victoria, Australia
- Department of Geriatric Medicine, Peninsula Health, Melbourne, Victoria, Australia
| |
Collapse
|
181
|
Differential effects of chronic immunosuppression on behavioral, epigenetic, and Alzheimer's disease-associated markers in 3xTg-AD mice. ALZHEIMERS RESEARCH & THERAPY 2021; 13:30. [PMID: 33472690 PMCID: PMC7818784 DOI: 10.1186/s13195-020-00745-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 12/07/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Circulating autoantibodies and sex-dependent discrepancy in prevalence are unexplained phenomena of Alzheimer's disease (AD). Using the 3xTg-AD mouse model, we reported that adult males show early manifestations of systemic autoimmunity, increased emotional reactivity, enhanced expression of the histone variant macroH2A1 in the cerebral cortex, and loss of plaque/tangle pathology. Conversely, adult females display less severe autoimmunity and retain their AD-like phenotype. This study examines the link between immunity and other traits of the current 3xTg-AD model. METHODS Young 3xTg-AD and wild-type mice drank a sucrose-laced 0.4 mg/ml solution of the immunosuppressant cyclophosphamide on weekends for 5 months. After behavioral phenotyping at 2 and 6 months of age, we assessed organ mass, serologic markers of autoimmunity, molecular markers of early AD pathology, and expression of genes associated with neurodegeneration. RESULTS Chronic immunosuppression prevented hematocrit drop and reduced soluble Aβ in 3xTg-AD males while normalizing the expression of histone variant macroH2A1 in 3xTg-AD females. This treatment also reduced hepatosplenomegaly, lowered autoantibody levels, and increased the effector T cell population while decreasing the proportion of regulatory T cells in both sexes. Exposure to cyclophosphamide, however, neither prevented reduced brain mass and BDNF expression nor normalized increased tau and anxiety-related behaviors. CONCLUSION The results suggest that systemic autoimmunity increases soluble Aβ production and affects transcriptional regulation of macroH2A1 in a sex-related manner. Despite the complexity of multisystem interactions, 3xTg-AD mice can be a useful in vivo model for exploring the regulatory role of autoimmunity in the etiology of AD-like neurodegenerative disorders.
Collapse
|
182
|
Calsolaro V, Bottari M, Coppini G, Lemmi B, Monzani F. Endocrine dysfunction and cognitive impairment. Minerva Endocrinol (Torino) 2021; 46:335-349. [PMID: 33435644 DOI: 10.23736/s2724-6507.20.03295-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Dementia is a highly prevalent chronic disease among the older population, affecting more than 50 million people worldwide and representing a huge healthcare, social and economic burden. Dementia, and in particular Alzheimer's disease, prevalence is expected to raise within the next few years. Unfortunately, no disease-modifying therapies are available so far, despite a plethora of clinical trials targeting the hallmarks of Alzheimer's disease. Given these premises, it appears crucial to address not only the neuropathological correlates of the disease, but also the modifiable risk factors. Among them, evidence suggest a role of the endocrine system not only in the brain development, but also in the maintenance of its health, having neurotrophic, antioxidant and metabolic functions crucial for the cognitive abilities. This review focuses on the evidence evaluating the impact of the endocrine systems, in particular thyroid function, insulin resistance, parathyroid hormone, vitamin D and sexual hormones on cognitive status. Results from epidemiological, preclinical and some clinical studies demonstrated the link between thyroid, parathyroid hormone and vitamin D and cognitive status, between diabetes, and insulin resistance in particular, and dementia, between sexual and adrenal hormones, particularly estrogen variation at menopause, and cognitive decline. The growing interest on the modifiable risks factors of cognitive decline increased the knowledge about the complex interplay of endocrine systems and cognition, highlighting the need and the usefulness of a multidisciplinary approach to the prevention of a complex and devastating disease.
Collapse
Affiliation(s)
- Valeria Calsolaro
- Geriatrics Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Marina Bottari
- Geriatrics Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Giulia Coppini
- Geriatrics Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Bianca Lemmi
- Geriatrics Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Fabio Monzani
- Geriatrics Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy -
| |
Collapse
|
183
|
Shen LX, Yang YX, Kuo K, Li HQ, Chen SD, Chen KL, Dong Q, Tan L, Yu JT. Social Isolation, Social Interaction, and Alzheimer's Disease: A Mendelian Randomization Study. J Alzheimers Dis 2021; 80:665-672. [PMID: 33579855 DOI: 10.3233/jad-201442] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Social isolation and social interaction have been suggested to be associated with Alzheimer's disease. However, the causality cannot be unambiguously assessed as traditional epidemiological methods are easily subject to unmeasured confounders and potential bias. OBJECTIVE To examine bidirectional relationships between social isolation, social interaction, and Alzheimer's disease using Mendelian randomization method for assessing potential causal inference. METHODS This bidirectional two-sample Mendelian randomization study used independent genetic variants associated with social isolation and social interaction (n = 302,567-487,647), and Alzheimer's disease (n = 455,258). MR analyses were performed using the inverse-variance-weighted (IVW) as the main MR analytical method to estimate the causal effect. For sensitivity analyses, we applied weighted median, MR Egger to further assess the credibility of the causal effect. RESULTS Of the five types of social engagement examined in our study, only one showed evidence of an association with the risk of Alzheimer's disease. Attendance at a gym or sports club (IVW OR per SD change: 0.670; 95% CI: 0.463-0.970; p = 0.034) was inversely associated with the risk of Alzheimer's disease. We also found that AD may reduce the attendance at religious group (IVW OR per SD change: 1.017; 95% CI: 1.005-1.030; p = 0.004). CONCLUSION This study suggests that regular attendance at a gym or sports club is causally associated with reduced risk of Alzheimer's disease. Further studies are warranted to elucidate potential mechanisms.
Collapse
Affiliation(s)
- Ling-Xiao Shen
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu-Xiang Yang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Kevin Kuo
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hong-Qi Li
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shi-Dong Chen
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ke-Liang Chen
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
184
|
Toljan K, Homolak J. Circadian changes in Alzheimer's disease: Neurobiology, clinical problems, and therapeutic opportunities. HANDBOOK OF CLINICAL NEUROLOGY 2021; 179:285-300. [PMID: 34225969 DOI: 10.1016/b978-0-12-819975-6.00018-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The understanding of Alzheimer's disease (AD) pathophysiology is an active area of research, and the traditional focus on hippocampus, amyloid and tau protein, and memory impairment has been expanded with components like neuroinflammation, insulin resistance, and circadian rhythm alterations. The bidirectional vicious cycle of neuroinflammation and neurodegeneration on a molecular level may cause functional deficits already long before the appearance of overt clinical symptoms. Located at the crossroads of metabolic, circadian, and hormonal signaling, the hypothalamus has been identified as another brain region affected by AD pathophysiology. Current findings on hypothalamic dysfunction open a broader horizon for studying AD pathogenesis and offer new opportunities for diagnosis and therapy. While treatments with cholinomimetics and memantine form a first line of pharmacological treatment, additional innovative research is pursued toward the development of antiinflammatory, growth factor, or antidiabetic types of medication. Following recent epidemiological data showing associations of AD incidence with modern societal and "life-style"-related risk factors, also nonpharmacological interventions, including sleep optimization, are being developed and some have been shown to be beneficial. Circadian aspects in AD are relevant from a pathophysiological standpoint, but they can also have an important role in pharmacologic and nonpharmacologic interventions, and appropriate timing of sleep, meals, and medication may boost therapeutic efficacy.
Collapse
Affiliation(s)
- Karlo Toljan
- Department of Neurology, Neurological Institute, Cleveland Clinic, Cleveland, OH, United States.
| | - Jan Homolak
- Department of Pharmacology, and Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
185
|
Yuan J, Maserejian N, Liu Y, Devine S, Gillis C, Massaro J, Au R. Severity Distribution of Alzheimer's Disease Dementia and Mild Cognitive Impairment in the Framingham Heart Study. J Alzheimers Dis 2020; 79:807-817. [PMID: 33361590 DOI: 10.3233/jad-200786] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Studies providing Alzheimer's disease (AD) prevalence data have largely neglected to characterize the proportion of AD that is mild, moderate, or severe. Estimates of the severity distribution along the AD continuum, including the mild cognitive impairment (MCI) stage, are important to plan research and allocate future resources, particularly resources targeted at particular stages of disease. OBJECTIVE To characterize the distribution of severity of AD dementia and MCI among prevalent cases in the population-based Framingham Heart Study. METHODS Participants (aged 50-94) with prevalent MCI or AD dementia clinical syndrome were cross-sectionally selected from three time-windows of the population-based Framingham Heart Study in 2004-2005 (n = 381), 2006-2007 (n = 422), and 2008-2009 (n = 389). Summary estimates of the severity distribution were achieved by pooling results across time-windows. Diagnosis and severity were assessed by consensus dementia review. MCI-progressive was determined if the participant had documented progression to AD dementia clinical syndrome using longitudinal data. RESULTS Among AD dementia participants, the pooled percentages were 50.4%for mild, 30.3%for moderate, and 19.3%for severe. Among all MCI and AD participants, the pooled percentages were 29.5%, 19.6%, 25.7%, and 45.2%for MCI-not-progressive, MCI-progressive, mild AD dementia, and the combined group of MCI-progressive and mild AD dementia, respectively. Distributions by age and sex were presented. CONCLUSION The finding that half of the people living with AD have mild disease underscores the need for research and interventions to slow decline or prevent progression of this burdensome disease.
Collapse
Affiliation(s)
- Jing Yuan
- Department of Neurology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China.,Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | | | - Yulin Liu
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA.,Framingham Heart Study, Boston University School of Medicine, Boston, MA, USA
| | - Sherral Devine
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA.,Framingham Heart Study, Boston University School of Medicine, Boston, MA, USA
| | - Cai Gillis
- Department of Epidemiology, Biogen, Cambridge, MA, USA
| | - Joseph Massaro
- Framingham Heart Study, Boston University School of Medicine, Boston, MA, USA.,Biostatistics Department, Boston University School of Public Health, Boston, MA, USA
| | - Rhoda Au
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA.,Framingham Heart Study, Boston University School of Medicine, Boston, MA, USA.,Department of Neurology, Boston University School of Medicine, Boston, MA, USA.,Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| |
Collapse
|
186
|
Reinscheid F. A new proposal for the causative agent of the sporadic form of Alzheimer's disease. Med Hypotheses 2020; 146:110453. [PMID: 33373829 DOI: 10.1016/j.mehy.2020.110453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/07/2020] [Accepted: 12/07/2020] [Indexed: 10/22/2022]
Abstract
Group B streptococcus (Streptococcus agalactiae) is proposed as causative agent for the development of the sporadic form of Alzheimer's disease. Using a fibrinogen binding protein, aggregates are formed including A-beta. After triggering Alzheimer's disease by the bacterium, the next down-stream events mainly follow the well known so called A-beta hypothesis. The combination of the two hypotheses is able to explain a number of epidemiological and biochemial aspects of Alzheimer's disease.
Collapse
Affiliation(s)
- Frauke Reinscheid
- Institution: GoePharmResearch, Pfaffenstück 16, 37077 Göttingen, Germany.
| |
Collapse
|
187
|
Wong E, Frost GR, Li YM. γ-Secretase Modulatory Proteins: The Guiding Hand Behind the Running Scissors. Front Aging Neurosci 2020; 12:614690. [PMID: 33343338 PMCID: PMC7738330 DOI: 10.3389/fnagi.2020.614690] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/10/2020] [Indexed: 12/16/2022] Open
Abstract
Described as the "proteasome of the membrane" or the "scissors in the membrane," γ-secretase has notoriously complicated biology, and even after decades of research, the full extent of its regulatory mechanism remains unclear. γ-Secretase is an intramembrane aspartyl protease complex composed of four obligatory subunits: Nicastrin (NCT), Presenilin (PS), Presenilin Enhancer-2 (Pen-2), and Anterior pharynx-defective-1 (Aph-1). γ-Secretase cleaves numerous type 1 transmembrane substrates, with no apparent homology, and plays major roles in broad biological pathways such as development, neurogenesis, and cancer. Notch and the amyloid precursor protein (APP) and are undoubtedly the best-studied γ-secretase substrates because of their role in cancer and Alzheimer's disease (AD) and therefore became the focus of increasing studies as an attractive therapeutic target. The regulation of γ-secretase is intricate and involves the function of multiple cellular entities. Recently, γ-secretase modulatory proteins (GSMPs), which are non-essential subunits and yet modulate γ-secretase activity and specificity, have emerged as an important component in guiding γ-secretase. GSMPs are responsive to cellular and environmental changes and therefore, provide another layer of regulation of γ-secretase. This type of enzymatic regulation allows for a rapid and fine-tuning of γ-secretase activity when appropriate signals appear enabling a temporal level of regulation. In this review article, we discuss the latest developments on GSMPs and implications on the development of effective therapeutics for γ-secretase-associated diseases such as AD and cancer.
Collapse
Affiliation(s)
- Eitan Wong
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | | | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
188
|
Alhazzani AA, Alqahtani AM, Alqahtani MS, Alahmari TM, Zarbah AA. Public awareness, knowledge, and attitude toward Alzheimer’s disease in Aseer region, Saudi Arabia. THE EGYPTIAN JOURNAL OF NEUROLOGY, PSYCHIATRY AND NEUROSURGERY 2020. [DOI: 10.1186/s41983-020-00213-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Alzheimer’s disease (AD) is a major public health problem and represents commonest form of dementia that affects many aspects of brain functions and contributes to 60–70% of dementia cases. No study has been conducted in Saudi Arabia to measure the prevalence of AD. However, approximately there are at least 50,000 patients in Saudi Arabia based on an expert’s estimation. Public knowledge and awareness about Alzheimer’s disease are crucial for early diagnosis and management.
Objective
To investigate the public’s knowledge and attitudes toward Alzheimer’s disease in Aseer region.
Subject and methods
Cross-sectional study has employed an electronic online, semi-structured, and self-administered questionnaire, randomly distributed among participants. The questions included sociodemographics, attitudes, and knowledge panels toward Alzheimer’s disease based on Alzheimer’s Disease Knowledge Scale (ADKS).
Results
The study included 1374 participants with a mean age 33 ± 11 years (ages18–70). The majority of the participants (77%) had no family history with Alzheimer’s disease. Less than 50% of the questions were answered correctly and the mean knowledge score was 10.77 ± 5.11. The results revealed that young-aged participants, females, and those having a family history of the disease, have significantly (P < 0.05) better AD-related knowledge.
Conclusion
The study revealed an inadequate AD-related knowledge as indicated by the low mean knowledge score. However, participants have demonstrated a positive attitude toward AD patients. More educational programs are required to enhance their awareness and knowledge about AD.
Collapse
|
189
|
Holingue C, Budavari AC, Rodriguez KM, Zisman CR, Windheim G, Fallin MD. Sex Differences in the Gut-Brain Axis: Implications for Mental Health. Curr Psychiatry Rep 2020; 22:83. [PMID: 33216233 PMCID: PMC7717677 DOI: 10.1007/s11920-020-01202-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/20/2020] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW The purpose of this article is to highlight how sex differences in the gut-brain axis may contribute to the discrepancies in incidence of neurodevelopmental, psychiatric, and neurodegenerative disorders between females and males. We focus on autism spectrum disorder, psychotic disorders, stress and anxiety disorders, depression, Alzheimer's disease, and Parkinson's disease and additionally discuss the comorbidity between inflammatory bowel disorder and mental health disorders. RECENT FINDINGS Human and animal studies show that sex may modify the relationship between the gut or immune system and brain and behavior. Sex also appears to modify the effect of microbial treatments such as probiotics and antibiotics on brain and behavior. There is emerging evidence that assessing the role of sex in the gut-brain axis may help elucidate the etiology of and identify effective treatments for neurodevelopmental, psychiatric, and neurodegenerative disorders.
Collapse
Affiliation(s)
- Calliope Holingue
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA.
- Department of Neuropsychology, Kennedy Krieger Institute, Baltimore, MD, USA.
- , Baltimore, USA.
| | - Alexa Curhan Budavari
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Katrina M Rodriguez
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Corina R Zisman
- Section on the Neurobiology of Fear and Anxiety, National Institute of Mental Health, Bethesda, MD, USA
| | - Grace Windheim
- Public Health Studies, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - M Daniele Fallin
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| |
Collapse
|
190
|
Lindbergh CA, Casaletto KB, Staffaroni AM, La Joie R, Iaccarino L, Edwards L, Tsoy E, Elahi F, Walters SM, Cotter D, You M, Apple AC, Asken B, Neuhaus J, Rexach JE, Wojta KJ, Rabinovici G, Kramer JH. Sex-related differences in the relationship between β-amyloid and cognitive trajectories in older adults. Neuropsychology 2020; 34:835-850. [PMID: 33030915 PMCID: PMC7839841 DOI: 10.1037/neu0000696] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Objective: We aimed to test the hypothesis that elevated neocortical β-amyloid (Aβ), a hallmark feature of Alzheimer's disease (AD), predicts sex-specific cognitive trajectories in clinically normal older adults, with women showing greater risk of decline than men. Method: Florbetapir Aβ positron emission tomography (PET) was acquired in 149 clinically normal older adults (52% female, Mage = 74). Participants underwent cognitive testing at baseline and during annual follow-up visits over a timespan of up to 5.14 years. Mixed-effects regression models evaluated whether relations between baseline neocortical Standardized Uptake Value Ratio (SUVR) and composite scores of episodic memory, executive functioning, and processing speed were moderated by sex (male/female) and apolipoprotein E (APOE) status (ε4 carrier/noncarrier). Results: Higher baseline SUVR was associated with longitudinal decline in episodic memory in women (b = -1.32, p < .001) but not men (b = -0.30, p = .28). Female APOE ε4 carriers with elevated SUVR showed particularly precipitous declines in episodic memory (b = -4.33, p < .001) whereas other cognitive domains were spared. SUVR did not predict changes in executive functioning or processing speed, regardless of sex (ps >.63), though there was a main effect of SUVR on processing speed (b = 2.50, p = .003). Conclusions: Clinically normal women with elevated Aβ are more vulnerable to episodic memory decline than men. Understanding sex-related differences in AD, particularly in preclinical stages, is crucial for guiding precision medicine approaches to early detection and intervention. (PsycInfo Database Record (c) 2020 APA, all rights reserved).
Collapse
Affiliation(s)
- Cutter A. Lindbergh
- Memory and Aging Center, Department of Neurology, University of California San Francisco
| | - Kaitlin B. Casaletto
- Memory and Aging Center, Department of Neurology, University of California San Francisco
| | - Adam M. Staffaroni
- Memory and Aging Center, Department of Neurology, University of California San Francisco
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, University of California San Francisco
| | - Leonardo Iaccarino
- Memory and Aging Center, Department of Neurology, University of California San Francisco
| | - Lauren Edwards
- Memory and Aging Center, Department of Neurology, University of California San Francisco
| | - Elena Tsoy
- Memory and Aging Center, Department of Neurology, University of California San Francisco
| | - Fanny Elahi
- Memory and Aging Center, Department of Neurology, University of California San Francisco
| | - Samantha M. Walters
- Memory and Aging Center, Department of Neurology, University of California San Francisco
| | - Devyn Cotter
- Memory and Aging Center, Department of Neurology, University of California San Francisco
| | - Michelle You
- Memory and Aging Center, Department of Neurology, University of California San Francisco
| | - Alexandra C. Apple
- Memory and Aging Center, Department of Neurology, University of California San Francisco
| | - Breton Asken
- Memory and Aging Center, Department of Neurology, University of California San Francisco
| | - John Neuhaus
- Department of Epidemiology and Biostatistics, University of California San Francisco
| | - Jessica E. Rexach
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California Los Angeles
| | - Kevin J. Wojta
- Department of Psychiatry, David Geffen School of Medicine, University of California Los Angeles
| | - Gil Rabinovici
- Memory and Aging Center, Department of Neurology, University of California San Francisco
| | - Joel H. Kramer
- Memory and Aging Center, Department of Neurology, University of California San Francisco
| | | |
Collapse
|
191
|
Umberson D, Donnelly R, Xu M, Farina M, Garcia MA. Death of a Child Prior to Midlife, Dementia Risk, and Racial Disparities. J Gerontol B Psychol Sci Soc Sci 2020; 75:1983-1995. [PMID: 31760426 PMCID: PMC7566971 DOI: 10.1093/geronb/gbz154] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES This study considers whether experiencing the death of a child prior to midlife (by parental age 40) is associated with subsequent dementia risk, and how such losses, which are more common for black than for white parents, may add to racial disparities in dementia risk. METHODS We use discrete-time event history models to predict dementia incidence among 9,276 non-Hispanic white and 2,182 non-Hispanic black respondents from the Health and Retirement Study, 2000-2014. RESULTS Losing a child prior to midlife is associated with increased risk for later dementia, and adds to disparities in dementia risk associated with race. The death of a child is associated with a number of biosocial variables that contribute to subsequent dementia risk, helping to explain how the death of child may increase risk over time. DISCUSSION The death of a child prior to midlife is a traumatic life course stressor with consequences that appear to increase dementia risk for both black and white parents, and this increased risk is explained by biosocial processes likely activated by bereavement. However, black parents are further disadvantaged in that they are more likely than white parents to experience the death of a child, and such losses add to the already substantial racial disadvantage in dementia risk.
Collapse
Affiliation(s)
- Debra Umberson
- Population Research Center, The University of Texas at Austin
- Department of Sociology, The University of Texas at Austin
| | - Rachel Donnelly
- Department of Sociology, Vanderbilt University, Nashville, Tennessee
| | - Minle Xu
- Population Research Center, The University of Texas at Austin
- Department of Sociology, The University of Texas at Austin
| | - Matthew Farina
- Population Research Center, The University of Texas at Austin
- Department of Sociology, The University of Texas at Austin
| | - Michael A Garcia
- Population Research Center, The University of Texas at Austin
- Department of Sociology, The University of Texas at Austin
| |
Collapse
|
192
|
Rouse HJ, Small BJ, Schinka JA, Hazlett AM, Loewenstein DA, Duara R, Potter H. Neuropsychiatric symptoms as a distinguishing factor between memory diagnoses. Int J Geriatr Psychiatry 2020; 35:1115-1122. [PMID: 32391573 DOI: 10.1002/gps.5333] [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] [Received: 04/10/2019] [Revised: 04/24/2020] [Accepted: 05/04/2020] [Indexed: 11/07/2022]
Abstract
OBJECTIVES To determine whether neuropsychiatric symptoms (NPS) are able to differentiate those with mild cognitive impairment (MCI) and dementia from persons who are cognitively healthy. METHODS Multinomial and binary logistic regressions were used to assess secondary data of a sample (n = 613) of older adults with NPS. Analyses evaluated the ability to differentiate between diagnoses, as well as the influence of these symptoms for individuals with amnestic MCI (MCI-A), non-amnestic MCI (MCI-NA), and dementia compared with those who are cognitively healthy. RESULTS Persons with MCI were more likely to have anxiety, apathy, and appetite changes compared with cognitively healthy individuals. Persons with dementia were more likely to have aberrant motor behaviors, anxiety, apathy, appetite changes, and delusions compared with those who were cognitively healthy. Individuals with any type of cognitive impairment were more likely to have anxiety, apathy, appetite changes, and delusions. Specifically, anxiety, apathy, appetite changes, and disinhibition were predictors of MCI-A; agitation and apathy were predictors of MCI-NA; and aberrant motor behaviors, anxiety, apathy, appetite changes, and delusions were predictors of dementia. Finally, nighttime behavior disorders were less likely in individuals with dementia. CONCLUSIONS The present study's results demonstrate that specific NPS are differentially represented among types of cognitive impairment and establish the predictive value for one of these cognitive impairment diagnoses.
Collapse
Affiliation(s)
- Hillary J Rouse
- School of Aging Studies, University of South Florida, Tampa, Florida, USA
| | - Brent J Small
- School of Aging Studies, University of South Florida, Tampa, Florida, USA
| | - John A Schinka
- School of Aging Studies, University of South Florida, Tampa, Florida, USA
| | - Abigail M Hazlett
- School of Aging Studies, University of South Florida, Tampa, Florida, USA
| | - David A Loewenstein
- Wien Center for Alzheimer's Disease and Memory Disorders, Mount Sinai Medical Center, University of Miami, Miami, Florida, USA
| | - Ranjan Duara
- Wien Center for Alzheimer's Disease and Memory Disorders, Mount Sinai Medical Center, University of Miami, Miami, Florida, USA
| | - Huntington Potter
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
193
|
Blood-brain barrier integrity in the pathogenesis of Alzheimer's disease. Front Neuroendocrinol 2020; 59:100857. [PMID: 32781194 DOI: 10.1016/j.yfrne.2020.100857] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 07/27/2020] [Accepted: 07/29/2020] [Indexed: 02/06/2023]
Abstract
The blood-brain barrier (BBB) tightly controls the molecular exchange between the brain parenchyma and blood. Accumulated evidence from transgenic animal Alzheimer's disease (AD) models and human AD patients have demonstrated that BBB dysfunction is a major player in AD pathology. In this review, we discuss the role of the BBB in maintaining brain integrity and how this is mediated by crosstalk between BBB-associated cells within the neurovascular unit (NVU). We then discuss the role of the NVU, in particular its endothelial cell, pericyte, and glial cell constituents, in AD pathogenesis. The effect of substances released by the neuroendocrine system in modulating BBB function and AD pathogenesis is also discussed. We perform a systematic review of currently available AD treatments specifically targeting pericytes and BBB glial cells. In summary, this review provides a comprehensive overview of BBB dysfunction in AD and a new perspective on the development of therapeutics for AD.
Collapse
|
194
|
Legdeur N, Tijms BM, Konijnenberg E, den Braber A, ten Kate M, Sudre CH, Tomassen J, Badissi M, Yaqub M, Barkhof F, van Berckel BN, Boomsma DI, Scheltens P, Holstege H, Maier AB, Visser PJ. Associations of Brain Pathology Cognitive and Physical Markers With Age in Cognitively Normal Individuals Aged 60-102 Years. J Gerontol A Biol Sci Med Sci 2020; 75:1609-1617. [PMID: 31411322 PMCID: PMC7494041 DOI: 10.1093/gerona/glz180] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Indexed: 01/23/2023] Open
Abstract
The prevalence of brain pathologies increases with age and cognitive and physical functions worsen over the lifetime. It is unclear whether these processes show a similar increase with age. We studied the association of markers for brain pathology cognitive and physical functions with age in 288 cognitively normal individuals aged 60-102 years selected from the cross-sectional EMIF-AD PreclinAD and 90+ Study at the Amsterdam UMC. An abnormal score was consistent with a score below the 5th percentile in the 60- to 70-year-old individuals. Prevalence of abnormal scores was estimated using Generalized Estimating Equations (GEE) models. The prevalence of abnormal handgrip strength, the Digit Symbol Substitution Test, and hippocampal volume showed the fastest increase with age and abnormal MMSE score, muscle mass, and amyloid aggregation the lowest. The increase in prevalence of abnormal markers was partly dependent on sex, level of education, and amyloid aggregation. We did not find a consistent pattern in which markers of brain pathology cognitive and physical processes became abnormal with age.
Collapse
Affiliation(s)
- Nienke Legdeur
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Betty M Tijms
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Elles Konijnenberg
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Anouk den Braber
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Mara ten Kate
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Carole H Sudre
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, UK
- Dementia Research Centre, Institute of Neurology, University College London, London, UK
- Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Jori Tomassen
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Maryam Badissi
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Maqsood Yaqub
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- European Society of Neuroradiology (ESNR), Institutes of Neurology and Healthcare Engineering, University College London, London, UK
| | - Bart N van Berckel
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Henne Holstege
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Clinical Genetics, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Andrea B Maier
- Department of Medicine and Aged Care, @AgeMelbourne, Royal Melbourne Hospital, University of Melbourne, Melbourne, Australia
- Department of Human Movement Sciences, @AgeAmsterdam, Vrije Universiteit Amsterdam, Research Institute Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Pieter Jelle Visser
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
195
|
Lai F, Mhatre PG, Yang Y, Wang M, Schupf N, Rosas HD. Sex differences in risk of Alzheimer's disease in adults with Down syndrome. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2020; 12:e12084. [PMID: 32995462 PMCID: PMC7507514 DOI: 10.1002/dad2.12084] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 07/11/2020] [Accepted: 07/13/2020] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Adults with Down syndrome (DS) older than 40 have Alzheimer's disease (AD) neuropathology and high risk for dementia, but little is known about the relationship of sex to AD risk in this population. METHODS Using nonparametric methods and Cox proportional hazards models we analyzed differences in incidence of dementia, by sex, presence of an apolipoprotein E (APOE) ε4 or ε2 allele, and dementia duration and decline in 246 adults over 40 with DS. RESULTS There was no significant sex difference in risk of AD or rate of cognitive decline. APOE ε4 allele significantly increased risk of AD irrespective of sex. No significant interactions were found between sex and APOE status on AD risk. Among those who died, dementia duration was significantly longer in women. DISCUSSION This study showed no effect of sex nor interaction between sex and APOE for risk of AD in adults with DS; however, women had longer dementia duration.
Collapse
Affiliation(s)
- Florence Lai
- Department of NeurologyMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Department of NeurologyMcLean HospitalBelmontMassachusettsUSA
| | - Pooja G. Mhatre
- Department of EpidemiologyMailman School of Public HealthColumbia UniversityNew YorkNew YorkUSA
| | - Yuchen Yang
- Department of BiostatisticsBloomberg School of Public HealthJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Mei‐Cheng Wang
- Department of BiostatisticsBloomberg School of Public HealthJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Nicole Schupf
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain and G.H. Sergievsky CenterColumbia UniversityNew YorkNew YorkUSA
- Departments of Neurology and PsychiatryColumbia University Medical CenterNew YorkNew YorkUSA
| | - H. Diana Rosas
- Department of NeurologyMassachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
- Department of NeurologyMcLean HospitalBelmontMassachusettsUSA
- Department of RadiologyAthinoula A. Martinos Center for Biomedical ImagingCharlestownMassachusettsUSA
| |
Collapse
|
196
|
Significance of Blood and Cerebrospinal Fluid Biomarkers for Alzheimer's Disease: Sensitivity, Specificity and Potential for Clinical Use. J Pers Med 2020; 10:jpm10030116. [PMID: 32911755 PMCID: PMC7565390 DOI: 10.3390/jpm10030116] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 08/21/2020] [Accepted: 09/01/2020] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia, affecting more than 5 million Americans, with steadily increasing mortality and incredible socio-economic burden. Not only have therapeutic efforts so far failed to reach significant efficacy, but the real pathogenesis of the disease is still obscure. The current theories are based on pathological findings of amyloid plaques and tau neurofibrillary tangles that accumulate in the brain parenchyma of affected patients. These findings have defined, together with the extensive neurodegeneration, the diagnostic criteria of the disease. The ability to detect changes in the levels of amyloid and tau in cerebrospinal fluid (CSF) first, and more recently in blood, has allowed us to use these biomarkers for the specific in-vivo diagnosis of AD in humans. Furthermore, other pathological elements of AD, such as the loss of neurons, inflammation and metabolic derangement, have translated to the definition of other CSF and blood biomarkers, which are not specific of the disease but, when combined with amyloid and tau, correlate with the progression from mild cognitive impairment to AD dementia, or identify patients who will develop AD pathology. In this review, we discuss the role of current and hypothetical biomarkers of Alzheimer's disease, their specificity, and the caveats of current high-sensitivity platforms for their peripheral detection.
Collapse
|
197
|
Gamache J, Yun Y, Chiba-Falek O. Sex-dependent effect of APOE on Alzheimer's disease and other age-related neurodegenerative disorders. Dis Model Mech 2020; 13:dmm045211. [PMID: 32859588 PMCID: PMC7473656 DOI: 10.1242/dmm.045211] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The importance of apolipoprotein E (APOE) in late-onset Alzheimer's disease (LOAD) has been firmly established, but the mechanisms through which it exerts its pathogenic effects remain elusive. In addition, the sex-dependent effects of APOE on LOAD risk and endophenotypes have yet to be explained. In this Review, we revisit the different aspects of APOE involvement in neurodegeneration and neurological diseases, with particular attention to sex differences in the contribution of APOE to LOAD susceptibility. We discuss the role of APOE in a broader range of age-related neurodegenerative diseases, and summarize the biological factors linking APOE to sex hormones, drawing on supportive findings from rodent models to identify major mechanistic themes underlying the exacerbation of LOAD-associated neurodegeneration and pathology in the female brain. Additionally, we list sex-by-genotype interactions identified across neurodegenerative diseases, proposing APOE variants as a shared etiology for sex differences in the manifestation of these diseases. Finally, we present recent advancements in 'omics' technologies, which provide a new platform for more in-depth investigations of how dysregulation of this gene affects the development and progression of neurodegenerative diseases. Collectively, the evidence summarized in this Review highlights the interplay between APOE and sex as a key factor in the etiology of LOAD and other age-related neurodegenerative diseases. We emphasize the importance of careful examination of sex as a contributing factor in studying the underpinning genetics of neurodegenerative diseases in general, but particularly for LOAD.
Collapse
Affiliation(s)
- Julia Gamache
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC 27708, USA
| | - Young Yun
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC 27708, USA
| | - Ornit Chiba-Falek
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC 27708, USA
| |
Collapse
|
198
|
Torromino G, Maggi A, De Leonibus E. Estrogen-dependent hippocampal wiring as a risk factor for age-related dementia in women. Prog Neurobiol 2020; 197:101895. [PMID: 32781107 DOI: 10.1016/j.pneurobio.2020.101895] [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: 12/20/2019] [Revised: 05/22/2020] [Accepted: 08/03/2020] [Indexed: 02/08/2023]
Abstract
Women are more prone than men to develop age-related dementia, such as Alzheimer's disease (AD). This has been linked to the marked decrease in circulating estrogens during menopause. This review proposes to change this perspective and consider women's vulnerability to developing AD as a consequence of sex differences in the neurobiology of memory, focusing on the hippocampus. The hippocampus of cognitively impaired subjects tends to shrink with age; however, in many cases, this can be prevented by exercise or cognitive training, suggesting that if you do not use the hippocampus you lose it. We will review the developmental trajectory of sex steroids-regulated differences on the hippocampus, proposing that the overall shaping action of sex-steroids results in a lower usage of the hippocampus in females, which in turn makes them more vulnerable to the effects of ageing, the "network fragility hypothesis". To explain why women rely less on hippocampus-dependent strategies, we propose a "computational hypothesis" that is based on experimental evidence suggesting that the direct effects of estrogens on hippocampal synaptic and structural plasticity during the estrous-cycle confers instability to the memory-dependent hippocampal network. Finally, we propose to counteract AD with training and/or treatments, such as orienteering, which specifically favour the use of the hippocampus.
Collapse
Affiliation(s)
- Giulia Torromino
- Telethon Institute of Genetics and Medicine (TIGEM), Telethon Foundation, Pozzuoli, Naples, Italy; Institute of Biochemistry and Cell Biology (IBBC), National Research Council, Monterotondo, Rome, Italy
| | - Adriana Maggi
- Center of Excellence on Neurodegenerative Diseases, University of Milan, Milan, Italy
| | - Elvira De Leonibus
- Telethon Institute of Genetics and Medicine (TIGEM), Telethon Foundation, Pozzuoli, Naples, Italy; Institute of Biochemistry and Cell Biology (IBBC), National Research Council, Monterotondo, Rome, Italy.
| |
Collapse
|
199
|
Sae-Lee W, Scott LL, Brose L, Encarnacion AJ, Shi T, Kore P, Oyibo LO, Ye C, Rozmiarek SK, Pierce JT. APP-Induced Patterned Neurodegeneration Is Exacerbated by APOE4 in Caenorhabditis elegans. G3 (BETHESDA, MD.) 2020; 10:2851-2861. [PMID: 32580938 PMCID: PMC7407474 DOI: 10.1534/g3.120.401486] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 06/19/2020] [Indexed: 11/18/2022]
Abstract
Genetic and epidemiological studies have found that variations in the amyloid precursor protein (APP) and the apoliopoprotein E (APOE) genes represent major modifiers of the progressive neurodegeneration in Alzheimer's disease (AD). An extra copy of or gain-of-function mutations in APP correlate with early onset AD. Compared to the other variants (APOE2 and APOE3), the ε4 allele of APOE (APOE4) hastens and exacerbates early and late onset forms of AD. Convenient in vivo models to study how APP and APOE4 interact at the cellular and molecular level to influence neurodegeneration are lacking. Here, we show that the nematode C. elegans can model important aspects of AD including age-related, patterned neurodegeneration that is exacerbated by APOE4 Specifically, we found that APOE4, but not APOE3, acts with APP to hasten and expand the pattern of cholinergic neurodegeneration caused by APP Molecular mechanisms underlying how APP and APOE4 synergize to kill some neurons while leaving others unaffected may be uncovered using this convenient worm model of neurodegeneration.
Collapse
Affiliation(s)
- Wisath Sae-Lee
- Center for Learning and Memory; Waggoner Center for Alcohol and Addiction Research; Cell and Molecular Biology; Department of Neuroscience, The University of Texas at Austin, TX, 78712
| | - Luisa L Scott
- Center for Learning and Memory; Waggoner Center for Alcohol and Addiction Research; Cell and Molecular Biology; Department of Neuroscience, The University of Texas at Austin, TX, 78712
| | - Lotti Brose
- Center for Learning and Memory; Waggoner Center for Alcohol and Addiction Research; Cell and Molecular Biology; Department of Neuroscience, The University of Texas at Austin, TX, 78712
| | - Aliyah J Encarnacion
- Center for Learning and Memory; Waggoner Center for Alcohol and Addiction Research; Cell and Molecular Biology; Department of Neuroscience, The University of Texas at Austin, TX, 78712
| | - Ted Shi
- Center for Learning and Memory; Waggoner Center for Alcohol and Addiction Research; Cell and Molecular Biology; Department of Neuroscience, The University of Texas at Austin, TX, 78712
| | - Pragati Kore
- Center for Learning and Memory; Waggoner Center for Alcohol and Addiction Research; Cell and Molecular Biology; Department of Neuroscience, The University of Texas at Austin, TX, 78712
| | - Lashaun O Oyibo
- Center for Learning and Memory; Waggoner Center for Alcohol and Addiction Research; Cell and Molecular Biology; Department of Neuroscience, The University of Texas at Austin, TX, 78712
| | - Congxi Ye
- Center for Learning and Memory; Waggoner Center for Alcohol and Addiction Research; Cell and Molecular Biology; Department of Neuroscience, The University of Texas at Austin, TX, 78712
| | - Susan K Rozmiarek
- Center for Learning and Memory; Waggoner Center for Alcohol and Addiction Research; Cell and Molecular Biology; Department of Neuroscience, The University of Texas at Austin, TX, 78712
| | - Jonathan T Pierce
- Center for Learning and Memory; Waggoner Center for Alcohol and Addiction Research; Cell and Molecular Biology; Department of Neuroscience, The University of Texas at Austin, TX, 78712
| |
Collapse
|
200
|
McFall GP, Bäckman L, Dixon RA. Nuances in Alzheimer's Genetic Risk Reveal Differential Predictions of Non-demented Memory Aging Trajectories: Selective Patterns by APOE Genotype and Sex. Curr Alzheimer Res 2020; 16:302-315. [PMID: 30873923 DOI: 10.2174/1567205016666190315094452] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/01/2019] [Accepted: 03/13/2019] [Indexed: 01/28/2023]
Abstract
BACKGROUND Apolipoprotein E (APOE) is a prominent genetic risk factor for Alzheimer's disease (AD) and a frequent target for associations with non-demented and cognitively impaired aging. APOE offers a unique opportunity to evaluate two dichotomous comparisons and selected gradations of APOE risk. Some evidence suggests that APOE effects may differ by sex and emerge especially in interaction with other AD-related biomarkers (e.g., vascular health). METHODS Longitudinal trajectories of non-demented adults (n = 632, 67% female, Mage = 68.9) populated a 40-year band of aging. Focusing on memory performance and individualized memory trajectories, a sequence of latent growth models was tested for predictions of (moderation between) APOE and pulse pressure (PP) as stratified by sex. The analyses (1) established robust benchmark PP effects on memory trajectories, (2) compared predictions of alternative dichotomous groupings (ε4- vs ε4+, ε2- vs ε2+), and (3) examined precision-based predictions by disaggregated APOE genotypes. RESULTS Healthier (lower) PP was associated with better memory performance and less decline. Therefore, all subsequent analyses were conducted in the interactive context of PP effects and sex stratification. The ε4-based dichotomization produced no differential genetic predictions. The ε2-based analyses showed sex differences, including selective protection for ε2-positive females. Exploratory follow-up disaggregated APOE genotype analyses suggested selective ε2 protection effects for both homozygotic and heterozygotic females. CONCLUSION Precision analyses of AD genetic risk will advance the understanding of underlying mechanisms and improve personalized implementation of interventions.
Collapse
Affiliation(s)
- G Peggy McFall
- Department of Psychology, University of Alberta, Edmonton, Canada.,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | | | - Roger A Dixon
- Department of Psychology, University of Alberta, Edmonton, Canada.,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| |
Collapse
|