1
|
Prajapati SK, Wang S, Mishra SP, Jain S, Yadav H. Protection of Alzheimer's disease progression by a human-origin probiotics cocktail. Sci Rep 2025; 15:1589. [PMID: 39794404 PMCID: PMC11724051 DOI: 10.1038/s41598-024-84780-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
Microbiome abnormalities (dysbiosis) significantly contribute to the progression of Alzheimer's disease (AD). However, the therapeutic efficacy of microbiome modulators in protecting against these ailments remains poorly studied. Herein, we tested a cocktail of unique probiotics, including 5 Lactobacillus and 5 Enterococcus strains isolated from infant gut with proven microbiome modulating capabilities. We aimed to determine the probiotics cocktail's efficacy in ameliorating AD pathology in a humanized AD mouse model of APP/PS1 strains. Remarkably, feeding mice with 1 × 1011 CFU per day in drinking water for 16 weeks significantly reduced cognitive decline (measured by the Morris Water Maze test) and AD pathology markers, such as Aβ aggregation, microglia activation, neuroinflammation, and preserved blood-brain barrier (BBB) tight junctions. The beneficial effects were linked to a reduced inflammatory microbiome, leading to decreased gut permeability and inflammation in both systemic circulation and the brain. Although both male and female mice showed overall improvements in cognition and biological markers, females did not exhibit improvements in specific markers related to inflammation and barrier permeability, suggesting that the underlying mechanisms may differ depending on sex. In conclusion, our results suggest that this unique probiotics cocktail could serve as a prophylactic agent to reduce the progression of cognitive decline and AD pathology. This is achieved by beneficially modulating the microbiome, improving intestinal tight junction proteins, reducing permeability in both gut and BBB, and decreasing inflammation in the gut, blood circulation, and brain, ultimately mitigating AD pathology and cognitive decline.
Collapse
Affiliation(s)
- Santosh Kumar Prajapati
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
- Department of Neurosurgery and Brain Repair, Center of Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Shaohua Wang
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
- Department of Neurosurgery and Brain Repair, Center of Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Department of Biomedical Sciences, Infectious and Tropical Disease Institute, Ohio University Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Sidharth P Mishra
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
- Department of Neurosurgery and Brain Repair, Center of Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Shalini Jain
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA
- Department of Neurosurgery and Brain Repair, Center of Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Hariom Yadav
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA.
- Department of Neurosurgery and Brain Repair, Center of Excellence in Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
- Department of Internal Medicine-Digestive Diseases and Nutrition, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
| |
Collapse
|
2
|
Verma S, Paliwal S, Paramanick D, Narayan CV, Saini M. Connecting the Dots: Gender, Sexuality, and Societal Influences on Cognitive Aging and Alzheimer's Disease. Curr Aging Sci 2025; 18:14-28. [PMID: 38899350 DOI: 10.2174/0118746098299754240530111755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/19/2024] [Accepted: 03/21/2024] [Indexed: 06/21/2024]
Abstract
Alzheimer's disease (AD) has many etiologies and the impact of gender on AD changes throughout time. As a consequence of advancements in precision medical procedures and methodology, Alzheimer's disease is now better understood and treated. Several risk factors may be addressed to lower one's chances of developing Alzheimer's disease or associated dementia (ADRD). The presence of amyloid-α protein senile plaques, intracellular tau protein neurofibrillary tangles (NfTs), neurodegeneration, and neuropsychiatric symptoms (NPS) characterizes Alzheimer's disease. NPS is common in persons with Alzheimer's disease dementia, although its presentation varies widely. Gender differences might explain this clinical variability. The fundamental goal of this review is to 1) emphasize the function of old age, sex, and gender in the development of Alzheimer's disease, dementia, and ADRD, and 2) explain the importance of sexual hormones, education, and APOE (Apolipoprotein E) status. This is a narrative summary of new ideas and concepts on the differences in the chance of developing dementia or Alzheimer's disease between men and women. A more thorough examination of risk and protective variables in both men and women might hasten research into the epidemiology of neurological illnesses such as dementia and Alzheimer's disease. Similarly, future preventive efforts should target men and women separately.
Collapse
Affiliation(s)
- Swati Verma
- Department of Pharmacy, ITS College of Pharmacy, Muradnagar, Ghaziabad, India
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India
| | - Sarvesh Paliwal
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India
| | - Debashish Paramanick
- Department of Pharmacy, School of Medical and Allied Science, KR Mangalam University, Gurugram, Haryana, India
| | | | - Manasvi Saini
- Department of Pharmacy, ITS College of Pharmacy, Muradnagar, Ghaziabad, India
| |
Collapse
|
3
|
Almutairi JA, Kidd EJ. Biological Sex Disparities in Alzheimer's Disease. Curr Top Behav Neurosci 2025; 69:79-104. [PMID: 39485650 DOI: 10.1007/7854_2024_545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Alzheimer's disease is a highly complex and multifactorial neurodegenerative disorder, with age being the most significant risk factor. The incidence of Alzheimer's disease doubles every 5 years after the age of 65. Consequently, one of the major challenges in Alzheimer's disease research is understanding how the brain changes with age. Gaining insights into these changes could help identify individuals who are more prone to developing Alzheimer's disease as they age. Over the past 25 years, studies on brain aging have examined thousands of human brains to explore the neuronal basis of age-related cognitive decline. However, most of these studies have focused on adults over 60, often neglecting the critical menopause transition period. During menopause, women experience a substantial decline in ovarian sex hormone production, with a decrease of about 90% in estrogen levels. Estrogen is known for its neuroprotective effects, and its significant loss during menopause affects various biological systems, including the brain. Importantly, despite known differences in dementia risk between sexes, the impact of biological sex and sex hormones on brain aging and the development of Alzheimer's disease remains underexplored.
Collapse
Affiliation(s)
- Jawza A Almutairi
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
- Department of Pharmaceutical Science, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Emma J Kidd
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK.
| |
Collapse
|
4
|
Yu G, Thorpe A, Zeng Q, Wang E, Cai D, Wang M, Zhang B. The Landscape of Sex- and APOE Genotype-Specific Transcriptional Changes in Alzheimer's Disease at the Single Cell Level. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.01.626234. [PMID: 39677706 PMCID: PMC11642736 DOI: 10.1101/2024.12.01.626234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, with approximately two-thirds of AD patients are females. Basic and clinical research studies show evidence supporting sex-specific differences contributing to the complexity of AD. There is also strong evidence supporting sex-specific interaction between the primary genetic risk factor of AD, APOE4 and AD-associated neurodegenerative processes. Recent studies by us and others have identified sex and/or APOE4 specific differentially expressed genes in AD based on the bulk tissue RNA-sequencing data of postmortem human brain samples in AD. However, there lacks a comprehensive investigation of the interplay between sex and APOE genotypes at the single cell level. In the current study, we systematically explore sex and APOE genotype differences in single cell transcriptomics in AD. Our work provides a comprehensive overview of sex and APOE genotype-specific transcriptomic changes across 54 high-resolution cell types in AD and highlights individual genes and brain cell types that show significant differences between sexes and APOE genotypes. This study lays the groundwork for exploring the complex molecular mechanisms of AD and will inform the development of effective sex- and APOE-stratified interventions for AD.
Collapse
|
5
|
López-Cerdán A, Andreu Z, Hidalgo MR, Soler-Sáez I, de la Iglesia-Vayá M, Mikozami A, Guerini FR, García-García F. An integrated approach to identifying sex-specific genes, transcription factors, and pathways relevant to Alzheimer's disease. Neurobiol Dis 2024; 199:106605. [PMID: 39009097 DOI: 10.1016/j.nbd.2024.106605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 06/06/2024] [Accepted: 07/11/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Age represents a significant risk factor for the development of Alzheimer's disease (AD); however, recent research has documented an influencing role of sex in several features of AD. Understanding the impact of sex on specific molecular mechanisms associated with AD remains a critical challenge to creating tailored therapeutic interventions. METHODS The exploration of the sex-based differential impact on disease (SDID) in AD used a systematic review to first select transcriptomic studies of AD with data regarding sex in the period covering 2002 to 2021 with a focus on the primary brain regions affected by AD - the cortex (CT) and the hippocampus (HP). A differential expression analysis for each study and two tissue-specific meta-analyses were then performed. Focusing on the CT due to the presence of significant SDID-related alterations, a comprehensive functional characterization was conducted: protein-protein network interaction and over-representation analyses to explore biological processes and pathways and a VIPER analysis to estimate transcription factor activity. RESULTS We selected 8 CT and 5 HP studies from the Gene Expression Omnibus (GEO) repository for tissue-specific meta-analyses. We detected 389 significantly altered genes in the SDID comparison in the CT. Generally, female AD patients displayed more affected genes than males; we grouped said genes into six subsets according to their expression profile in female and male AD patients. Only subset I (repressed genes in female AD patients) displayed significant results during functional profiling. Female AD patients demonstrated more significant impairments in biological processes related to the regulation and organization of synapsis and pathways linked to neurotransmitters (glutamate and GABA) and protein folding, Aβ aggregation, and accumulation compared to male AD patients. These findings could partly explain why we observe more pronounced cognitive decline in female AD patients. Finally, we detected 23 transcription factors with different activation patterns according to sex, with some associated with AD for the first time. All results generated during this study are readily available through an open web resource Metafun-AD (https://bioinfo.cipf.es/metafun-ad/). CONCLUSION Our meta-analyses indicate the existence of differences in AD-related mechanisms in female and male patients. These sex-based differences will represent the basis for new hypotheses and could significantly impact precision medicine and improve diagnosis and clinical outcomes in AD patients.
Collapse
Affiliation(s)
- Adolfo López-Cerdán
- Computational Biomedicine Laboratory, Principe Felipe Research Center (CIPF), 46012, Valencia, Spain; Biomedical Imaging Unit FISABIO-CIPF, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana, 46012, Valencia, Spain
| | - Zoraida Andreu
- Foundation Valencian Institute of Oncology (FIVO), 46009, Valencia, Spain
| | - Marta R Hidalgo
- Computational Biomedicine Laboratory, Principe Felipe Research Center (CIPF), 46012, Valencia, Spain
| | - Irene Soler-Sáez
- Computational Biomedicine Laboratory, Principe Felipe Research Center (CIPF), 46012, Valencia, Spain
| | - María de la Iglesia-Vayá
- Biomedical Imaging Unit FISABIO-CIPF, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana, 46012, Valencia, Spain
| | - Akiko Mikozami
- Oral Health/Brain Health/Total health (OBT) Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | | | - Francisco García-García
- Computational Biomedicine Laboratory, Principe Felipe Research Center (CIPF), 46012, Valencia, Spain.
| |
Collapse
|
6
|
Woodward M, Bennett DA, Rundek T, Perry G, Rudka T. The relationship between hippocampal changes in healthy aging and Alzheimer's disease: a systematic literature review. Front Aging Neurosci 2024; 16:1390574. [PMID: 39210976 PMCID: PMC11357962 DOI: 10.3389/fnagi.2024.1390574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction Neurobiological changes in the hippocampus are a common consequence of aging. However, there are differences in the rate of decline and overall volume loss in people with no cognitive impairment compared to those with mild cognitive impairment (MCI) and Alzheimer's disease (AD). This systematic literature review was conducted to determine the relationship between hippocampal atrophy and changes in hippocampal volume in the non-cognitively impaired brain and those with MCI or AD. Methods This systematic review was guided by the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) methodology. The PubMed database was searched up to September 15, 2022, for longitudinal magnetic resonance imaging studies reporting hippocampal atrophy or volume change in cognitively normal aging individuals and patients with MCI and/or AD. Study selection was divided into two steps: (1) identification and retrieval of relevant studies; (2) screening the studies by (a) title/abstract and (b) full text. Two teams, each consisting of two independent reviewers, determined whether the publications met the inclusion criteria for the systematic review. An evidence table was populated with data extracted from eligible publications and inclusion in the final systematic review was confirmed. Results The systematic search identified 357 publications that were initially screened by title/abstract, of which, 115 publications were retrieved and reviewed by full text for eligibility. Seventeen publications met the eligibility criteria; however, during data extraction, two studies were determined to not meet the inclusion criteria and were excluded. The remaining 15 studies were included in the systematic review. Overall, the results of these studies demonstrated that the hippocampus and hippocampal subfields change over time, with both decreased hippocampal volume and increased rate of hippocampal atrophy observed. Hippocampal changes in AD were observed to be greater than hippocampal changes in MCI, and changes in MCI were observed to be greater than those in normal aging populations. Conclusion Published literature suggests that the rate of hippocampal decline and extent of loss is on a continuum that begins in people without cognitive impairment and continues to MCI and AD, and that differences between no cognitive impairment, MCI, and AD are quantitative rather than qualitative.
Collapse
Affiliation(s)
- Michael Woodward
- Austin Health, University of Melbourne, Heidelberg, VIC, Australia
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, United States
| | - Tatjana Rundek
- Evelyn F. McKnight Brain Institute, Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - George Perry
- Department of Neuroscience, Developmental and Regenerative Biology, University of Texas at San Antonio, San Antonio, TX, United States
| | - Tomasz Rudka
- Danone Specialised Nutrition, Hoofddorp, Netherlands
| |
Collapse
|
7
|
Arenaza‐Urquijo EM, Boyle R, Casaletto K, Anstey KJ, Vila‐Castelar C, Colverson A, Palpatzis E, Eissman JM, Kheng Siang Ng T, Raghavan S, Akinci M, Vonk JMJ, Machado LS, Zanwar PP, Shrestha HL, Wagner M, Tamburin S, Sohrabi HR, Loi S, Bartrés‐Faz D, Dubal DB, Vemuri P, Okonkwo O, Hohman TJ, Ewers M, Buckley RF. Sex and gender differences in cognitive resilience to aging and Alzheimer's disease. Alzheimers Dement 2024; 20:5695-5719. [PMID: 38967222 PMCID: PMC11350140 DOI: 10.1002/alz.13844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 03/08/2024] [Accepted: 03/21/2024] [Indexed: 07/06/2024]
Abstract
Sex and gender-biological and social constructs-significantly impact the prevalence of protective and risk factors, influencing the burden of Alzheimer's disease (AD; amyloid beta and tau) and other pathologies (e.g., cerebrovascular disease) which ultimately shape cognitive trajectories. Understanding the interplay of these factors is central to understanding resilience and resistance mechanisms explaining maintained cognitive function and reduced pathology accumulation in aging and AD. In this narrative review, the ADDRESS! Special Interest Group (Alzheimer's Association) adopted a multidisciplinary approach to provide the foundations and recommendations for future research into sex- and gender-specific drivers of resilience, including a sex/gender-oriented review of risk factors, genetics, AD and non-AD pathologies, brain structure and function, and animal research. We urge the field to adopt a sex/gender-aware approach to resilience to advance our understanding of the intricate interplay of biological and social determinants and consider sex/gender-specific resilience throughout disease stages. HIGHLIGHTS: Sex differences in resilience to cognitive decline vary by age and cognitive status. Initial evidence supports sex-specific distinctions in brain pathology. Findings suggest sex differences in the impact of pathology on cognition. There is a sex-specific change in resilience in the transition to clinical stages. Gender and sex factors warrant study: modifiable, immune, inflammatory, and vascular.
Collapse
Affiliation(s)
- Eider M. Arenaza‐Urquijo
- Environment and Health Over the Life Course Programme, Climate, Air Pollution, Nature and Urban Health ProgrammeBarcelona Institute for Global Health (ISGlobal)BarcelonaSpain
- University of Pompeu FabraBarcelonaBarcelonaSpain
| | - Rory Boyle
- Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Kaitlin Casaletto
- Department of NeurologyMemory and Aging CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Kaarin J. Anstey
- University of New South Wales Ageing Futures InstituteSydneyNew South WalesAustralia
- Neuroscience Research AustraliaSydneyNew South WalesAustralia
- School of Psychology, University of New South WalesSidneyNew South WalesAustralia
| | | | - Aaron Colverson
- University of Florida Center for Arts in Medicine Interdisciplinary Research LabUniversity of Florida, Center of Arts in MedicineGainesvilleFloridaUSA
| | - Eleni Palpatzis
- Environment and Health Over the Life Course Programme, Climate, Air Pollution, Nature and Urban Health ProgrammeBarcelona Institute for Global Health (ISGlobal)BarcelonaSpain
- University of Pompeu FabraBarcelonaBarcelonaSpain
| | - Jaclyn M. Eissman
- Vanderbilt Memory and Alzheimer's Center, Department of NeurologyVanderbilt University Medical CenterNashvilleTennesseeUSA
- Vanderbilt Genetics InstituteVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Ted Kheng Siang Ng
- Rush Institute for Healthy Aging and Department of Internal MedicineRush University Medical CenterChicagoIllinoisUSA
| | | | - Muge Akinci
- Environment and Health Over the Life Course Programme, Climate, Air Pollution, Nature and Urban Health ProgrammeBarcelona Institute for Global Health (ISGlobal)BarcelonaSpain
- University of Pompeu FabraBarcelonaBarcelonaSpain
| | - Jet M. J. Vonk
- Department of NeurologyMemory and Aging CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Luiza S. Machado
- Graduate Program in Biological Sciences: Biochemistry, Universidade Federal Do Rio Grande Do Sul, FarroupilhaPorto AlegreBrazil
| | - Preeti P. Zanwar
- Jefferson College of Population Health, Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
- The Network on Life Course and Health Dynamics and Disparities, University of Southern CaliforniaLos AngelesCaliforniaUSA
| | | | - Maude Wagner
- Rush Alzheimer's Disease Center, Rush University Medical CenterChicagoIllinoisUSA
| | - Stefano Tamburin
- Department of Neurosciences, Biomedicine and Movement SciencesUniversity of VeronaVeronaItaly
| | - Hamid R. Sohrabi
- Centre for Healthy AgeingHealth Future InstituteMurdoch UniversityMurdochWestern AustraliaAustralia
- School of Psychology, Murdoch UniversityMurdochWestern AustraliaAustralia
| | - Samantha Loi
- Neuropsychiatry Centre, Royal Melbourne HospitalParkvilleVictoriaAustralia
- Department of PsychiatryUniversity of MelbourneParkvilleVictoriaAustralia
| | - David Bartrés‐Faz
- Department of MedicineFaculty of Medicine and Health Sciences & Institut de NeurociènciesUniversity of BarcelonaBarcelonaBarcelonaSpain
- Institut d'Investigacions Biomèdiques (IDIBAPS)BarcelonaBarcelonaSpain
- Institut Guttmann, Institut Universitari de Neurorehabilitació adscrit a la Universitat Autónoma de BarcelonaBadalonaBarcelonaSpain
| | - Dena B. Dubal
- Department of Neurology and Weill Institute of NeurosciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Biomedical and Neurosciences Graduate ProgramsUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | | | - Ozioma Okonkwo
- Alzheimer's Disease Research Center and Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Timothy J. Hohman
- Vanderbilt Memory and Alzheimer's Center, Department of NeurologyVanderbilt University Medical CenterNashvilleTennesseeUSA
- Vanderbilt Genetics InstituteVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Michael Ewers
- Institute for Stroke and Dementia ResearchKlinikum der Universität MünchenLudwig Maximilians Universität (LMU)MunichGermany
- German Center for Neurodegenerative Diseases (DZNE, Munich)MunichGermany
| | - Rachel F. Buckley
- Massachusetts General HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | | |
Collapse
|
8
|
Zhao Y, Caffo BS, Luo X. Longitudinal regression of covariance matrix outcomes. Biostatistics 2024; 25:385-401. [PMID: 36451549 PMCID: PMC11650757 DOI: 10.1093/biostatistics/kxac045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 10/25/2022] [Accepted: 11/02/2022] [Indexed: 02/17/2024] Open
Abstract
In this study, a longitudinal regression model for covariance matrix outcomes is introduced. The proposal considers a multilevel generalized linear model for regressing covariance matrices on (time-varying) predictors. This model simultaneously identifies covariate-associated components from covariance matrices, estimates regression coefficients, and captures the within-subject variation in the covariance matrices. Optimal estimators are proposed for both low-dimensional and high-dimensional cases by maximizing the (approximated) hierarchical-likelihood function. These estimators are proved to be asymptotically consistent, where the proposed covariance matrix estimator is the most efficient under the low-dimensional case and achieves the uniformly minimum quadratic loss among all linear combinations of the identity matrix and the sample covariance matrix under the high-dimensional case. Through extensive simulation studies, the proposed approach achieves good performance in identifying the covariate-related components and estimating the model parameters. Applying to a longitudinal resting-state functional magnetic resonance imaging data set from the Alzheimer's Disease (AD) Neuroimaging Initiative, the proposed approach identifies brain networks that demonstrate the difference between males and females at different disease stages. The findings are in line with existing knowledge of AD and the method improves the statistical power over the analysis of cross-sectional data.
Collapse
Affiliation(s)
- Yi Zhao
- Department of Biostatistics and Health Data Science, Indiana University
School of Medicine, 410 W 10th Street, Indianapolis, IN 46202, USA
| | - Brian S Caffo
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public
Health, 615 N Wolfe Street, Baltimore, MD 21205, USA
| | - Xi Luo
- Department of Biostatistics and Data Science, The University of Texas
Health Science Center at Houston, 1200 Pressler Street, Houston, TX
77030, USA
| |
Collapse
|
9
|
Wagemann O, Li Y, Hassenstab J, Aschenbrenner AJ, McKay NS, Gordon BA, Benzinger TLS, Xiong C, Cruchaga C, Renton AE, Perrin RJ, Berman SB, Chhatwal JP, Farlow MR, Day GS, Ikeuchi T, Jucker M, Lopera F, Mori H, Noble JM, Sánchez‐Valle R, Schofield PR, Morris JC, Daniels A, Levin J, Bateman RJ, McDade E, Llibre‐Guerra JJ. Investigation of sex differences in mutation carriers of the Dominantly Inherited Alzheimer Network. Alzheimers Dement 2024; 20:47-62. [PMID: 37740921 PMCID: PMC10841236 DOI: 10.1002/alz.13460] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/10/2023] [Accepted: 08/13/2023] [Indexed: 09/25/2023]
Abstract
INTRODUCTION Studies suggest distinct differences in the development, presentation, progression, and response to treatment of Alzheimer's disease (AD) between females and males. We investigated sex differences in cognition, neuroimaging, and fluid biomarkers in dominantly inherited AD (DIAD). METHODS Three hundred twenty-five mutation carriers (55% female) and one hundred eighty-six non-carriers (58% female) of the Dominantly Inherited Alzheimer Network Observational Study were analyzed. Linear mixed models and Spearman's correlation explored cross-sectional sex differences in cognition, cerebrospinal fluid (CSF) biomarkers, Pittsburgh compound B positron emission tomography (11 C-PiB PET) and structural magnetic resonance imaging (MRI). RESULTS Female carriers performed better than males on delayed recall and processing speed despite similar hippocampal volumes. As the disease progressed, symptomatic females revealed higher increases in MRI markers of neurodegeneration and memory impairment. PiB PET and established CSF AD markers revealed no sex differences. DISCUSSION Our findings suggest an initial cognitive reserve in female carriers followed by a pronounced increase in neurodegeneration coupled with worse performance on delayed recall at later stages of DIAD.
Collapse
Affiliation(s)
- Olivia Wagemann
- Department of NeurologyWashington University St. LouisSt. LouisMissouriUSA
- Department of NeurologyLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Yan Li
- Department of BiostatisticsWashington University St. LouisSt. LouisMissouriUSA
| | - Jason Hassenstab
- Department of NeurologyWashington University St. LouisSt. LouisMissouriUSA
| | | | - Nicole S. McKay
- Department of RadiologyWashington University St. LouisSt. LouisMissouriUSA
| | - Brian A. Gordon
- Department of RadiologyWashington University St. LouisSt. LouisMissouriUSA
| | | | - Chengjie Xiong
- Department of BiostatisticsWashington University St. LouisSt. LouisMissouriUSA
| | - Carlos Cruchaga
- Department of PsychiatryWashington University St. LouisSt. LouisMissouriUSA
| | - Alan E. Renton
- Department of NeuroscienceIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Richard J. Perrin
- Department of NeurologyWashington University St. LouisSt. LouisMissouriUSA
- Department of Pathology and ImmunologyWashington University St. LouisSt. LouisMissouriUSA
| | - Sarah B. Berman
- Department of NeurologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Jasmeer P. Chhatwal
- Department of NeurologyMassachusetts General and Brigham & Female's HospitalsHarvard Medical SchoolBostonMassachusettsUSA
| | - Martin R. Farlow
- Department of NeurologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Gregory S. Day
- Department of NeurologyMayo Clinic FloridaJacksonvilleFloridaUSA
| | - Takeshi Ikeuchi
- Department of Molecular GeneticsBrain Research InstituteNiigata UniversityNiigataJapan
| | - Mathias Jucker
- Hertie Institute for Clinical Brain ResearchUniversity of TübingenTübingenGermany
- German Center for Neurodegenerative Diseases (DZNE)TübingenGermany
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia (GNA)Universidad de AntioquiaMedellinColombia
| | - Hiroshi Mori
- Department of Clinical NeuroscienceOsaka Metropolitan University Medical SchoolNagaoka Sutoku UniversityOsakaJapan
| | - James M. Noble
- Department of NeurologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Raquel Sánchez‐Valle
- Department of NeurologyHospital Clínic de Barcelona (IDIBAPS)University of BarcelonaBarcelonaSpain
| | - Peter R. Schofield
- Neuroscience Research AustraliaSydneyNew South WalesAustralia
- School of Biomedical SciencesUniversity of New South WalesSydneyNew South WalesAustralia
| | - John C. Morris
- Department of NeurologyWashington University St. LouisSt. LouisMissouriUSA
| | - Alisha Daniels
- Department of NeurologyWashington University St. LouisSt. LouisMissouriUSA
| | - Johannes Levin
- Department of NeurologyLudwig‐Maximilians‐Universität MünchenMunichGermany
- German Center for Neurodegenerative Diseases (DZNE)MunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - Randall J. Bateman
- Department of NeurologyWashington University St. LouisSt. LouisMissouriUSA
| | - Eric McDade
- Department of NeurologyWashington University St. LouisSt. LouisMissouriUSA
| | | | | |
Collapse
|
10
|
Lin X, Feng T, Cui E, Li Y, Qin Z, Zhao X. A rat model established by simulating genetic-environmental interactions recapitulates human Alzheimer's disease pathology. Brain Res 2024; 1822:148663. [PMID: 37918702 DOI: 10.1016/j.brainres.2023.148663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/16/2023] [Accepted: 10/30/2023] [Indexed: 11/04/2023]
Abstract
BACKGROUND In humans, Alzheimer's disease (AD) is typically sporadic in nature, and its pathology is usually influenced by extensive factors. The study established a rat model based on the genetic-environmental interaction. METHODS A rat model was established by transduction of an adeno-associated virus combined with acrolein treatment. Rats were assigned to the normal control (NC), acrolein group, AAV (-) group, AAV-APP group, and AAV-APP/acrolein group. The success of model construction was verified in multiple ways, including by assessing cognitive function, examining microstructural changes in the brain in vivo, and performing immunohistochemistry. The contribution of genetic (APP mutation) and environmental (acrolein) factors to AD-like phenotypes in the model was explored by factorial analysis. RESULTS 1) The AAV-APP/acrolein group showed a decline in cognitive function, as indicated by a reduced gray matter volume in key cognition-related brain areas, lower FA values in the hippocampus and internal olfactory cortex, and Aβ deposition in the cortex and hippocampus. 2) The AAV-APP group also showed a decline in cognitive function, although the group exhibited atypical brain atrophy in the gray matter and insignificant Aβ deposition. 3) The acrolein group did not show any significant changes in Aβ levels, gray matter volume, or cognitive function. 4) The genetic factor (APP mutation) explained 39.74% of the AD-like phenotypes in the model factors, and the environmental factor (acrolein exposure) explained 33.3%. CONCLUSIONS The genetic-environmental interaction rat model exhibited a phenotype that resembled the features of human AD and will be useful for research on AD.
Collapse
Affiliation(s)
- Xiaomei Lin
- Department of Imaging, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200000, China
| | - Tianyuyi Feng
- Department of Imaging, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200000, China
| | - Erheng Cui
- Department of Imaging, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200000, China
| | - Yunfei Li
- Department of Imaging, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200000, China
| | - Zhang Qin
- Department of Imaging, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200000, China
| | - Xiaohu Zhao
- Department of Imaging, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200000, China.
| |
Collapse
|
11
|
Sauty B, Durrleman S. Impact of sex and APOE- ε4 genotype on patterns of regional brain atrophy in Alzheimer's disease and healthy aging. Front Neurol 2023; 14:1161527. [PMID: 37333001 PMCID: PMC10272760 DOI: 10.3389/fneur.2023.1161527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 05/19/2023] [Indexed: 06/20/2023] Open
Abstract
Alzheimer's Disease (AD) is a heterogeneous disease that disproportionately affects women and people with the APOE-ε4 susceptibility gene. We aim to describe the not-well-understood influence of both risk factors on the dynamics of brain atrophy in AD and healthy aging. Regional cortical thinning and brain atrophy were modeled over time using non-linear mixed-effect models and the FreeSurfer software with t1-MRI scans from the Alzheimer's Disease Neuroimaging Initiative (N = 1,502 subjects, 6,728 images in total). Covariance analysis was used to disentangle the effect of sex and APOE genotype on the regional onset age and pace of atrophy, while correcting for educational level. A map of the regions mostly affected by neurodegeneration is provided. Results were confirmed on gray matter density data from the SPM software. Women experience faster atrophic rates in the temporal, frontal, parietal lobes and limbic system and earlier onset in the amygdalas, but slightly later onset in the postcentral and cingulate gyri as well as all regions of the basal ganglia and thalamus. APOE-ε4 genotypes leads to earlier and faster atrophy in the temporal, frontal, parietal lobes, and limbic system in AD patients, but not in healthy patients. Higher education was found to slightly delay atrophy in healthy patients, but not for AD patients. A cohort of amyloid positive patients with MCI showed a similar impact of sex as in the healthy cohort, while APOE-ε4 showed similar associations as in the AD cohort. Female sex is as strong a risk factor for AD as APOE-ε4 genotype regarding neurodegeneration. Women experience a sharper atrophy in the later stages of the disease, although not a significantly earlier onset. These findings may have important implications for the development of targeted intervention.
Collapse
|
12
|
Klingenberg M, Stark D, Eitel F, Budding C, Habes M, Ritter K. Higher performance for women than men in MRI-based Alzheimer's disease detection. Alzheimers Res Ther 2023; 15:84. [PMID: 37081528 PMCID: PMC10116672 DOI: 10.1186/s13195-023-01225-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 04/03/2023] [Indexed: 04/22/2023]
Abstract
INTRODUCTION Although machine learning classifiers have been frequently used to detect Alzheimer's disease (AD) based on structural brain MRI data, potential bias with respect to sex and age has not yet been addressed. Here, we examine a state-of-the-art AD classifier for potential sex and age bias even in the case of balanced training data. METHODS Based on an age- and sex-balanced cohort of 432 subjects (306 healthy controls, 126 subjects with AD) extracted from the ADNI data base, we trained a convolutional neural network to detect AD in MRI brain scans and performed ten different random training-validation-test splits to increase robustness of the results. Classifier decisions for single subjects were explained using layer-wise relevance propagation. RESULTS The classifier performed significantly better for women (balanced accuracy [Formula: see text]) than for men ([Formula: see text]). No significant differences were found in clinical AD scores, ruling out a disparity in disease severity as a cause for the performance difference. Analysis of the explanations revealed a larger variance in regional brain areas for male subjects compared to female subjects. DISCUSSION The identified sex differences cannot be attributed to an imbalanced training dataset and therefore point to the importance of examining and reporting classifier performance across population subgroups to increase transparency and algorithmic fairness. Collecting more data especially among underrepresented subgroups and balancing the dataset are important but do not always guarantee a fair outcome.
Collapse
Affiliation(s)
- Malte Klingenberg
- Charité - Universitätsmedizin Berlin (corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health), Department of Psychiatry and Neurosciences, Berlin, Germany
- Bernstein Center for Computational Neuroscience, Berlin, Germany
| | - Didem Stark
- Charité - Universitätsmedizin Berlin (corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health), Department of Psychiatry and Neurosciences, Berlin, Germany
- Bernstein Center for Computational Neuroscience, Berlin, Germany
| | - Fabian Eitel
- Charité - Universitätsmedizin Berlin (corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health), Department of Psychiatry and Neurosciences, Berlin, Germany
- Bernstein Center for Computational Neuroscience, Berlin, Germany
| | - Céline Budding
- Eindhoven University of Technology, Eindhoven, Netherlands
| | - Mohamad Habes
- Neuroimage Analytics Laboratory and Biggs Institute Neuroimaging Core, Glenn Biggs Institute for Neurodegenerative Disorders, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Kerstin Ritter
- Charité - Universitätsmedizin Berlin (corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health), Department of Psychiatry and Neurosciences, Berlin, Germany.
- Bernstein Center for Computational Neuroscience, Berlin, Germany.
| |
Collapse
|
13
|
Li H, Huang Z, Gao Z, Zhu W, Li Y, Zhou S, Li X, Yu Y. Sex Difference in General Cognition Associated with Coupling of Whole-brain Functional Connectivity Strength to Cerebral Blood Flow Changes During Alzheimer's Disease Progression. Neuroscience 2023; 509:187-200. [PMID: 36496188 DOI: 10.1016/j.neuroscience.2022.12.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 11/28/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a progressive age-related neurodegenerative disorder that results in irreversible cognitive impairments. Nonetheless, there are numerous sex-dependent differences in clinical course. We examined potential contributions of neurovascular coupling deficits to sex differences in AD progression. T1-weighted three-dimensional structural magnetic resonance images, functional blood oxygen level dependent and arterial spin labeling images were acquired from 50 AD patients (28 females), 52 amnesic mild cognitive impairment patients (31 females), and 59 healthy controls (36 females). Short- and long-range functional connectivity strength (FCS) and cerebral blood flow (CBF) values were calculated for all participants. Then, the CBF/FCS coupling ratio, which represented the amount of blood supply per unit of connectivity strength, was calculated for each voxel. Two-way ANOVA was performed to identify group × sex interactions and main effects of group. Correlation analysis was used to assess associations between CBF/FCS ratios and Mini-Mental State Examination (MMSE). There were significant group × sex interaction effects on short-range coupling ratios of right middle temporal gyrus, left angular gyrus, left inferior orbital frontal gyrus, and left superior frontal gyrus as well as on the long-range coupling ratios of right middle temporal gyrus, left precuneus, left posterior cingulate cortex, and left angular gyrus. There were significant negative correlations between MMSE scores and CBF/FCS ratios for all regions with significant group × sex interactions among female patients, while positive correlations were found among male patients. Our results demonstrate significant sex differences in neurovascular coupling mechanisms associated with cognitive function during the course of AD.
Collapse
Affiliation(s)
- Hui Li
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Ziang Huang
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Ziwen Gao
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Wanqiu Zhu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yuqing Li
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Shanshan Zhou
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Xiaoshu Li
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| | - Yongqiang Yu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| |
Collapse
|
14
|
Stites SD, Cao H, James R, Harkins K, Coykendall C, Flatt JD. A systematic review of measures of gender and biological sex: Exploring candidates for Alzheimer's disease and related dementias (AD/ADRD) research. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2023; 15:e12359. [PMID: 36845632 PMCID: PMC9943901 DOI: 10.1002/dad2.12359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/19/2022] [Accepted: 08/23/2022] [Indexed: 02/24/2023]
Abstract
Background Gender and biological sex are social and structural determinants of health and umbrella concepts encompassing many distinct attributes. This systematic review summarizes measures of gender and biological sex published in the biomedical literature. The goal was to identify measures that may be useful to researchers studying Alzheimer's disease and Alzheimer's disease related dementias (AD/ADRD). Methods A search of PubMed, Embase, and PsycINFO (ProQuest platform) databases from 2000 to 2021 identified 1454 articles, which were then screened by five independent reviewers. Measures of gender and biological sex are summarized according to theoretical commitments and psychometric properties. Results Twenty-nine measures were identified that assessed gender-related constructs, and 4 were identified that assessed biological factors. Self-report instruments characterized aspects of gender, such as gender stereotypes, norms, and ideologies. One measure was developed with a focus on older adults (65+ years). Discussion We offer recommendations to guide measurement of gender in AD/ADRD research, including how the use of specific existing measures may help advance AD/ADRD research. The lack of gender measures for older adults limits AD/ADRD research. New measures may be needed to address lifespan and generational differences in gender factors. Highlights A review of articles identifies 29 measures of gender in biomedical research.Gender is captured using multidimensional, self-reported concepts.One measure was developed with a focus on older adults (65+).
Collapse
Affiliation(s)
- Shana D. Stites
- Department of PsychiatryPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Hannah Cao
- School of Social Policy and PracticeUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Richard James
- University of Pennsylvania LibrariesPhiladelphiaPennsylvaniaUSA
| | - Kristin Harkins
- Division of Geriatric MedicineDepartment of MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Cameron Coykendall
- Division of Geriatric MedicineDepartment of MedicinePerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Jason D. Flatt
- Department of Social and Behavioral HealthSchool of Public HealthUniversity of NevadaLas VegasNevadaUSA
| |
Collapse
|
15
|
Santiago JA, Potashkin JA. Biological and Clinical Implications of Sex-Specific Differences in Alzheimer's Disease. Handb Exp Pharmacol 2023; 282:181-197. [PMID: 37460661 DOI: 10.1007/164_2023_672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
Mounting evidence indicates that the female sex is a risk factor for Alzheimer's disease (AD), the most common cause of dementia worldwide. Decades of research suggest that sex-specific differences in genetics, environmental factors, hormones, comorbidities, and brain structure and function may contribute to AD development. However, although significant progress has been made in uncovering specific genetic factors and biological pathways, the precise mechanisms underlying sex-biased differences are not fully characterized. Here, we review several lines of evidence, including epidemiological, clinical, and molecular studies addressing sex differences in AD. In addition, we discuss the challenges and future directions in advancing personalized treatments for AD.
Collapse
Affiliation(s)
| | - Judith A Potashkin
- Cellular and Molecular Pharmacology Department, Center for Neurodegenerative Diseases and Therapeutics, The Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA.
| |
Collapse
|
16
|
Jett S, Dyke JP, Andy C, Schelbaum E, Jang G, Boneu Yepez C, Pahlajani S, Diaz I, Diaz Brinton R, Mosconi L. Sex and menopause impact 31P-Magnetic Resonance Spectroscopy brain mitochondrial function in association with 11C-PiB PET amyloid-beta load. Sci Rep 2022; 12:22087. [PMID: 36543814 PMCID: PMC9772209 DOI: 10.1038/s41598-022-26573-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Increasing evidence implicates sex and endocrine aging effects on brain bioenergetic aging in the greater lifetime risk of Alzheimer's disease (AD) in women. We conducted 31Phosphorus Magnetic Resonance Spectroscopy (31P-MRS) to assess the impact of sex and menopause on brain high-energy phosphates [adenosine triphosphate (ATP), phosphocreatine (PCr), inorganic phosphate (Pi)] and membrane phospholipids [phosphomonoesters/phosphodiesters (PME/PDE)] in 216 midlife cognitively normal individuals at risk for AD, 80% female. Ninety-seven participants completed amyloid-beta (Aβ) 11C-PiB PET. Women exhibited higher ATP utilization than men in AD-vulnerable frontal, posterior cingulate, fusiform, medial and lateral temporal regions (p < 0.001). This profile was evident in frontal cortex at the pre-menopausal and peri-menopausal stage and extended to the other regions at the post-menopausal stage (p = 0.001). Results were significant after multi-variable adjustment for age, APOE-4 status, midlife health indicators, history of hysterectomy/oophorectomy, use of menopause hormonal therapy, and total intracranial volume. While associations between ATP/PCr and Aβ load were not significant, individuals with the highest Aβ load were post-menopausal and peri-menopausal women with ATP/PCr ratios in the higher end of the distribution. No differences in Pi/PCr, Pi/ATP or PME/PDE were detected. Outcomes are consistent with dynamic bioenergetic brain adaptations that are associated with female sex and endocrine aging.
Collapse
Affiliation(s)
- Steven Jett
- Department of Neurology, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Jonathan P Dyke
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Caroline Andy
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Eva Schelbaum
- Department of Neurology, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Grace Jang
- Department of Neurology, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Camila Boneu Yepez
- Department of Neurology, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Silky Pahlajani
- Department of Neurology, Weill Cornell Medicine, New York, NY, 10021, USA
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Ivan Diaz
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Roberta Diaz Brinton
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
- Department of Neurology, University of Arizona, Tucson, AZ, USA
| | - Lisa Mosconi
- Department of Neurology, Weill Cornell Medicine, New York, NY, 10021, USA.
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
17
|
Taylor A, Zhang F, Niu X, Heywood A, Stocks J, Feng G, Popuri K, Beg MF, Wang L. Investigating the temporal pattern of neuroimaging-based brain age estimation as a biomarker for Alzheimer's Disease related neurodegeneration. Neuroimage 2022; 263:119621. [PMID: 36089183 PMCID: PMC9995621 DOI: 10.1016/j.neuroimage.2022.119621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/29/2022] [Accepted: 09/07/2022] [Indexed: 11/19/2022] Open
Abstract
Neuroimaging-based brain-age estimation via machine learning has emerged as an important new approach for studying brain aging. The difference between one's estimated brain age and chronological age, the brain age gap (BAG), has been proposed as an Alzheimer's Disease (AD) biomarker. However, most past studies on the BAG have been cross-sectional. Quantifying longitudinal changes in an individual's BAG temporal pattern would likely improve prediction of AD progression and clinical outcome based on neurophysiological changes. To fill this gap, our study conducted predictive modeling using a large neuroimaging dataset with up to 8 years of follow-up to examine the temporal patterns of the BAG's trajectory and how it varies by subject-level characteristics (sex, APOEɛ4 carriership) and disease status. Specifically, we explored the pattern and rate of change in BAG over time in individuals who remain stable with normal cognition or mild cognitive impairment (MCI), as well as individuals who progress to clinical AD. Combining multimodal imaging data in a support vector regression model to estimate brain age yielded improved performance over single modality. Multilevel modeling results showed the BAG followed a linear increasing trajectory with a significantly faster rate in individuals with MCI who progressed to AD compared to cognitively normal or MCI individuals who did not progress. The dynamic changes in the BAG during AD progression were further moderated by sex and APOEɛ4 carriership. Our findings demonstrate the BAG as a potential biomarker for understanding individual specific temporal patterns related to AD progression.
Collapse
Affiliation(s)
- Alexei Taylor
- Department of Psychological and Brain Sciences, Drexel University, Philadelphia, PA 19104, USA
| | - Fengqing Zhang
- Department of Psychological and Brain Sciences, Drexel University, Philadelphia, PA 19104, USA.
| | - Xin Niu
- Department of Psychological and Brain Sciences, Drexel University, Philadelphia, PA 19104, USA
| | - Ashley Heywood
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jane Stocks
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Gangyi Feng
- Department of Linguistics and Modern Languages, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China; Brain and Mind Institute, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Karteek Popuri
- Department of Computer Science, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Mirza Faisal Beg
- School of Engineering Science, Simon Fraser University, Burnaby, V6A1S6 BCE, Canada
| | - Lei Wang
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Psychiatry and Behavioral Health, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
18
|
Chang YL, Moscovitch M. Sex differences in item and associative memory among older adults with amnestic mild cognitive impairment. Neuropsychologia 2022; 176:108375. [PMID: 36179862 DOI: 10.1016/j.neuropsychologia.2022.108375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 09/01/2022] [Accepted: 09/22/2022] [Indexed: 10/31/2022]
Abstract
In older adults without cognitive impairment, women have an advantage over men in verbal memory tests; however, whether women with amnestic mild cognitive impairment (aMCI) exhibit this advantage remains controversial. We evaluated sex-specific differences in older adults with and without aMCI in item and associative verbal memory by using an associative memory task with immediate and delayed recognition conditions. The associations between memory task performances and medial temporal morphometric measures were examined. The study included 49 individuals with aMCI and 55 healthy older adults (HOs). The results revealed that a female advantage in immediate item and delayed associative memory was evident in HOs, and the female advantage in associative memory persisted even after item memory performance was controlled. By contrast, the female advantage was absent in individuals with aMCI; such women had more associative false alarms than men with aMCI. Furthermore, decreases in item memory, associative memory, and cortical thickness in the perirhinal and entorhinal regions in individuals with aMCI versus their sex-matched controls were more prominent in women than in men. The relation between brain structure and associative memory function was evident only for women, indicating that women and men may have different cognitive and neural mechanisms for processing associative memory. These findings support the concept of cognitive reserve in women during normal aging. Accounting for sex differences in verbal memory performance is crucial to improve aMCI identification, particularly for women.
Collapse
Affiliation(s)
- Yu-Ling Chang
- Department of Psychology, College of Science, National Taiwan University, Taipei, 10617, Taiwan; Neurobiology and Cognitive Science Center, National Taiwan University, Taipei, 10617, Taiwan; Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, 10048, Taiwan; Center for Artificial Intelligence and Advanced Robotics, National Taiwan University, Taipei, 10617, Taiwan.
| | - Morris Moscovitch
- Rotman Research Institute, Baycrest Health Sciences, Toronto, ON M6A 2E1, Canada; Department of Psychology, University of Toronto, Toronto, ON M5S 3G3, Canada
| |
Collapse
|
19
|
Contador J, Pérez-Millan A, Guillen N, Sarto J, Tort-Merino A, Balasa M, Falgàs N, Castellví M, Borrego-Écija S, Juncà-Parella J, Bosch B, Fernández-Villullas G, Ramos-Campoy O, Antonell A, Bargalló N, Sanchez-Valle R, Sala Llonch R, Lladó A. Sex differences in early-onset Alzheimer's disease. Eur J Neurol 2022; 29:3623-3632. [PMID: 36005384 DOI: 10.1111/ene.15531] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 08/10/2022] [Accepted: 08/13/2022] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Sex is believed to drive heterogeneity in Alzheimer's disease (AD), although evidence in early-onset AD (<65 years, EOAD) is scarce. METHODS We included 62 EOAD patients and 44 healthy controls (HC) with cerebrospinal fluid (CSF) AD's core biomarkers and neurofilament light chain levels, neuropsychological assessment, and 3T-MRI. We measured cortical thickness (CTh) and hippocampal subfield volumes (HpS) using Freesurfer. Adjusted linear models were used to analyze sex-differences and the relationship between atrophy and cognition. RESULTS Compared to same-sex HC, female-EOAD showed greater cognitive impairment and broader atrophy burden than male-EOAD. In a direct female-EOAD and male-EOAD comparison, there were slight differences in temporal CTh, with no differences in cognition or HpS. CSF tau levels were higher in female-EOAD than in male-EOAD. Greater atrophy was associated with worse cognition in female-EOAD. CONCLUSIONS At diagnosis, there are sex-differences in the pattern of cognitive impairment, atrophy burden and CSF tau in EOAD, suggesting there is an influence of sex on pathology spreading and susceptibility to the disease in EOAD.
Collapse
Affiliation(s)
- José Contador
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Agnès Pérez-Millan
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain.,Institute of Neurosciences. Department of Biomedicine, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Nuria Guillen
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Jordi Sarto
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Adrià Tort-Merino
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Mircea Balasa
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain.,Atlantic Fellow for Equity in Brain Health, Global Brain Heath Institute
| | - Neus Falgàs
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain.,Atlantic Fellow for Equity in Brain Health, Global Brain Heath Institute
| | - Magdalena Castellví
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Sergi Borrego-Écija
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Jordi Juncà-Parella
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Beatriz Bosch
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Guadalupe Fernández-Villullas
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Oscar Ramos-Campoy
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Anna Antonell
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Nuria Bargalló
- Image Diagnostic Centre Radiology Department, Hospital Clínic de Barcelona, Magnetic Resonance Image Core facility Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Salud Mental. CIBERSAM., Spain
| | - Raquel Sanchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Roser Sala Llonch
- Institute of Neurosciences. Department of Biomedicine, Faculty of Medicine, University of Barcelona, Barcelona, Spain.,Biomedical Imaging Group, Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain
| | - Albert Lladó
- Alzheimer's Disease and Other Cognitive Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
20
|
Sex differences in risk factors that predict progression from mild cognitive impairment to Alzheimer's dementia. J Int Neuropsychol Soc 2022; 29:360-368. [PMID: 35968841 DOI: 10.1017/s1355617722000297] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVES To evaluate whether cerebrospinal fluid biomarkers, apolipoprotein e4, neuroimaging abnormalities, and neuropsychological data differentially predict progression from mild cognitive impairment (MCI) to dementia for men and women. METHODS Participants who were diagnosed with MCI at baseline (n = 449) were classified as either progressing to Alzheimer's dementia at follow-up or as not progressing. Men and women were first compared using bivariate analyses. Sex-stratified Cox proportional hazard regressions were performed examining the relationship between baseline data and the likelihood of progressing to dementia. Sex interactions were subsequently examined. RESULTS Cox proportional hazard regression controlling for age and education indicated that all variables significantly predicted subsequent progression to dementia for men and women. Sex interactions indicated that only Rey Auditory Verbal Learning Test (RAVLT) delayed recall and Functional Activities Questionnaire (FAQ) were significantly stronger risk factors for women. When all variables were entered into a fully adjusted model, significant risk factors for women were Aβ42, hippocampal volume, RAVLT delayed recall, Boston Naming Test, and FAQ. In contrast, for men, Aβ42, p-tau181, p-tau181/Aβ42, hippocampal volume, category fluency and FAQ were significant risk factors. Interactions with sex were only significant for p-tau181/Aβ42 and RAVLT delayed recall for the fully adjusted model. CONCLUSIONS Men and women with MCI may to differ for which factors predict subsequent dementia although future analyses with greater power are needed to evaluate sex differences. We hypothesize that brain and cognitive reserve theories may partially explain these findings.
Collapse
|
21
|
Buckley RF, O'Donnell A, McGrath ER, Jacobs HI, Lois C, Satizabal CL, Ghosh S, Rubinstein ZB, Murabito JM, Sperling RA, Johnson KA, Seshadri S, Beiser AS. Menopause Status Moderates Sex Differences in Tau Burden: A Framingham PET Study. Ann Neurol 2022; 92:11-22. [PMID: 35471588 PMCID: PMC9233144 DOI: 10.1002/ana.26382] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Women have a higher lifetime risk of Alzheimer's disease (AD) than men. Among cognitively normal (CN) older adults, women exhibit elevated tau positron emission tomography (PET) signal compared with men. We explored whether menopause exacerbates sex differences in tau deposition in middle-aged adults. METHODS 328 CN participants from the Framingham Study (mean age = 57 years (±10 years), 161 women, of whom, 104 were post-menopausal) underwent tau and β-amyloid (Aβ)-PET neuroimaging. We examined global Aβ-PET, and tau-PET signal in 5 regions identified a priori as demonstrating significant sex differences in older adults (in temporal, inferior parietal, middle frontal, and lateral occipital regions). We examined sex and menopause status-related differences in each region-of-interest, using linear regressions, as well as interactions with Aβ and APOEε4 genotype. RESULTS Women exhibited higher tau-PET signal (p < 0.002), and global Aβ-PET (p = 0.010), than men in inferior parietal, rostral middle frontal, and lateral occipital regions. Compared with age-matched men, post-menopausal women showed significantly higher tau-PET signal in parieto-occipital regions (p < 0.0001). By contrast, no differences in tau-PET signal existed between pre-menopausal women and men. Aβ-PET was not associated with menopausal status or age. Neither Aβ-PET nor APOEε4 status moderated sex or menopause associations with tau-PET. INTERPRETATION Clear divergence in tauopathy between the sexes are apparent approximately 20 years earlier than previously reported. Menopause status moderated sex differences in Aβ and tau-PET burden, with tau first appearing post-menopause. Sex and menopause differences consistently appeared in middle frontal and parieto-occipital regions but were not moderated by Aβ burden or APOEε4, suggesting that menopause-related tau vulnerability may be independent of AD-related pathways. ANN NEUROL 2022;92:11-22.
Collapse
Affiliation(s)
- Rachel F. Buckley
- Department of NeurologyMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
- Center for Alzheimer Research and Treatment, Department of NeurologyBrigham and Women's HospitalBostonMAUSA
- Melbourne School of Psychological Science and Florey InstitutesUniversity of MelbourneParkvilleVICAustralia
| | - Adrienne O'Donnell
- Department of BiostatisticsBoston University School of Public HealthBostonMAUSA
- Framingham Heart StudyFraminghamMAUSA
| | - Emer R. McGrath
- Framingham Heart StudyFraminghamMAUSA
- HRB Clinical Research FacilityNational University of Ireland GalwayGalwayIreland
| | - Heidi I.L. Jacobs
- Gordon Center for Medical Imaging, Department of RadiologyMassachusetts General Hospital/Harvard Medical SchoolBostonMAUSA
- Faculty of Health, Medicine and Life Sciences, School for Mental Health and Neuroscience, Alzheimer Centre LimburgMaastricht UniversityMaastrichtThe Netherlands
| | - Cristina Lois
- Gordon Center for Medical Imaging, Department of RadiologyMassachusetts General Hospital/Harvard Medical SchoolBostonMAUSA
| | - Claudia L. Satizabal
- Framingham Heart StudyFraminghamMAUSA
- Glen Biggs Institute for Alzheimer's & Neurodegenerative DiseasesUniversity of Texas Health San AntonioSan AntonioTXUSA
- Department of NeurologyBoston University School of MedicineBostonMAUSA
| | | | - Zoe B. Rubinstein
- Gordon Center for Medical Imaging, Department of RadiologyMassachusetts General Hospital/Harvard Medical SchoolBostonMAUSA
| | | | - Reisa A. Sperling
- Department of NeurologyMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
- Center for Alzheimer Research and Treatment, Department of NeurologyBrigham and Women's HospitalBostonMAUSA
| | - Keith A. Johnson
- Center for Alzheimer Research and Treatment, Department of NeurologyBrigham and Women's HospitalBostonMAUSA
- Gordon Center for Medical Imaging, Department of RadiologyMassachusetts General Hospital/Harvard Medical SchoolBostonMAUSA
| | - Sudha Seshadri
- Framingham Heart StudyFraminghamMAUSA
- Glen Biggs Institute for Alzheimer's & Neurodegenerative DiseasesUniversity of Texas Health San AntonioSan AntonioTXUSA
- Department of NeurologyBoston University School of MedicineBostonMAUSA
| | - Alexandra S. Beiser
- Department of BiostatisticsBoston University School of Public HealthBostonMAUSA
- Framingham Heart StudyFraminghamMAUSA
- Department of NeurologyBoston University School of MedicineBostonMAUSA
| |
Collapse
|
22
|
Lai Z, Zhang Q, Liang L, Wei Y, Duan G, Mai W, Zhao L, Liu P, Deng D. Efficacy and Mechanism of Moxibustion Treatment on Mild Cognitive Impairment Patients: An fMRI Study Using ALFF. Front Mol Neurosci 2022; 15:852882. [PMID: 35620445 PMCID: PMC9127659 DOI: 10.3389/fnmol.2022.852882] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Background Mild Cognitive Impairment (MCI), as a high risk of Alzheimer’s disease (AD), represents a state of cognitive function between normal aging and dementia. Moxibustion may effectively delay the progression of AD, while there is a lack of studies on the treatments in MCI. This study aimed to evaluate the effect of moxibustion treatment revealed by the amplitude of low-frequency fluctuation (ALFF) in MCI. Method We enrolled 30 MCI patients and 30 matched healthy controls (HCs) in this study. We used ALFF to compare the difference between MCI and HCs at baseline and the regulation of spontaneous neural activity in MCI patients by moxibustion. The Mini-Mental State Examination and Montreal Cognitive Assessment scores were used to evaluate cognitive function. Results Compared with HCs, the ALFF values significantly decreased in the right temporal poles: middle temporal gyrus (TPOmid), right inferior temporal gyrus, left middle cingulate gyrus, and increased in the left hippocampus, left middle temporal gyrus, right lingual gyrus, and right middle occipital gyrus in MCI patients. After moxibustion treatment, the ALFF values notably increased in the left precuneus, left thalamus, right temporal poles: middle temporal gyrus, right middle frontal gyrus, right inferior temporal gyrus, right putamen, right hippocampus, and right fusiform gyrus, while decreased in the bilateral lingual gyrus in MCI patients. The Mini-Mental State Examination and Montreal Cognitive Assessment scores increased after moxibustion treatment, and the increase in Mini-Mental State Examination score was positively correlated with the increase of ALFF value in the right TPOmid, the right insula, and the left superior temporal gyrus. Conclusion Moxibustion treatment might improve the cognitive function of MCI patients by modulating the brain activities within the default mode network, visual network, and subcortical network with a trend of increased ALFF values and functional asymmetry of the hippocampus. These results indicate that moxibustion holds great potential in the treatment of MCI.
Collapse
Affiliation(s)
- Ziyan Lai
- Department of Radiology, The People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, China
| | - Qingping Zhang
- Department of Radiology, The People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, China
| | - Lingyan Liang
- Department of Radiology, The People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, China
| | - Yichen Wei
- Department of Radiology, The People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, China
| | - Gaoxiong Duan
- Department of Radiology, The People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, China
| | - Wei Mai
- Department of Acupuncture, The First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, China
| | - Lihua Zhao
- Department of Acupuncture, The First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, China
| | - Peng Liu
- Life Science Research Center, School of Life Science and Technology, Xidian University, Xi’an, China
| | - Demao Deng
- Department of Radiology, The People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, China
- *Correspondence: Demao Deng
| |
Collapse
|
23
|
Abstract
Sex and gender differences are seen in cognitive disturbances in a variety of neurological and psychiatry diseases. Men are more likely to have cognitive symptoms in schizophrenia whereas women are more likely to have more severe cognitive symptoms with major depressive disorder and Alzheimer's disease. Thus, it is important to understand sex and gender differences in underlying cognitive abilities with and without disease. Sex differences are noted in performance across various cognitive domains - with males typically outperforming females in spatial tasks and females typically outperforming males in verbal tasks. Furthermore, there are striking sex differences in brain networks that are activated during cognitive tasks and in learning strategies. Although rarely studied, there are also sex differences in the trajectory of cognitive aging. It is important to pay attention to these sex differences as they inform researchers of potential differences in resilience to age-related cognitive decline and underlying mechanisms for both healthy and pathological cognitive aging, depending on sex. We review literature on the progressive neurodegenerative disorder, Alzheimer's disease, as an example of pathological cognitive aging in which human females show greater lifetime risk, neuropathology, and cognitive impairment, compared to human males. Not surprisingly, the relationships between sex and cognition, cognitive aging, and Alzheimer's disease are nuanced and multifaceted. As such, this chapter will end with a discussion of lifestyle factors, like education and diet, as modifiable factors that can alter cognitive aging by sex. Understanding how cognition changes across age and contributing factors, like sex differences, will be essential to improving care for older adults.
Collapse
|
24
|
Jett S, Malviya N, Schelbaum E, Jang G, Jahan E, Clancy K, Hristov H, Pahlajani S, Niotis K, Loeb-Zeitlin S, Havryliuk Y, Isaacson R, Brinton RD, Mosconi L. Endogenous and Exogenous Estrogen Exposures: How Women's Reproductive Health Can Drive Brain Aging and Inform Alzheimer's Prevention. Front Aging Neurosci 2022; 14:831807. [PMID: 35356299 PMCID: PMC8959926 DOI: 10.3389/fnagi.2022.831807] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/07/2022] [Indexed: 01/14/2023] Open
Abstract
After advanced age, female sex is the major risk factor for late-onset Alzheimer's disease (AD), the most common cause of dementia affecting over 24 million people worldwide. The prevalence of AD is higher in women than in men, with postmenopausal women accounting for over 60% of all those affected. While most research has focused on gender-combined risk, emerging data indicate sex and gender differences in AD pathophysiology, onset, and progression, which may help account for the higher prevalence in women. Notably, AD-related brain changes develop during a 10-20 year prodromal phase originating in midlife, thus proximate with the hormonal transitions of endocrine aging characteristic of the menopause transition in women. Preclinical evidence for neuroprotective effects of gonadal sex steroid hormones, especially 17β-estradiol, strongly argue for associations between female fertility, reproductive history, and AD risk. The level of gonadal hormones to which the female brain is exposed changes considerably across the lifespan, with relevance to AD risk. However, the neurobiological consequences of hormonal fluctuations, as well as that of hormone therapies, are yet to be fully understood. Epidemiological studies have yielded contrasting results of protective, deleterious and null effects of estrogen exposure on dementia risk. In contrast, brain imaging studies provide encouraging evidence for positive associations between greater cumulative lifetime estrogen exposure and lower AD risk in women, whereas estrogen deprivation is associated with negative consequences on brain structure, function, and biochemistry. Herein, we review the existing literature and evaluate the strength of observed associations between female-specific reproductive health factors and AD risk in women, with a focus on the role of endogenous and exogenous estrogen exposures as a key underlying mechanism. Chief among these variables are reproductive lifespan, menopause status, type of menopause (spontaneous vs. induced), number of pregnancies, and exposure to hormonal therapy, including hormonal contraceptives, hormonal therapy for menopause, and anti-estrogen treatment. As aging is the greatest risk factor for AD followed by female sex, understanding sex-specific biological pathways through which reproductive history modulates brain aging is crucial to inform preventative and therapeutic strategies for AD.
Collapse
Affiliation(s)
- Steven Jett
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Niharika Malviya
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Eva Schelbaum
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Grace Jang
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Eva Jahan
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Katherine Clancy
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Hollie Hristov
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Silky Pahlajani
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
- Department of Radiology, Weill Cornell Medical College, New York, NY, United States
| | - Kellyann Niotis
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Susan Loeb-Zeitlin
- Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY, United States
| | - Yelena Havryliuk
- Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY, United States
| | - Richard Isaacson
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Roberta Diaz Brinton
- Department of Pharmacology, University of Arizona, Tucson, AZ, United States
- Department of Neurology, University of Arizona, Tucson, AZ, United States
| | - Lisa Mosconi
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
- Department of Radiology, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
25
|
Cieri F, Zhuang X, Cordes D, Kaplan N, Cummings J, Caldwell J. Relationship of sex differences in cortical thickness and memory among cognitively healthy subjects and individuals with mild cognitive impairment and Alzheimer disease. Alzheimers Res Ther 2022; 14:36. [PMID: 35193682 PMCID: PMC8864917 DOI: 10.1186/s13195-022-00973-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 01/27/2022] [Indexed: 12/22/2022]
Abstract
BACKGROUND An aging society has increased rates of late onset Alzheimer disease dementia (ADD), the most common form of age-related dementia. This neurodegenerative disease disproportionately affects women. METHODS We use data from the Alzheimer's Disease Neuroimaging Initiative (ADNI) to examine sex differences in cortical thickness (CT) and memory performance. Analyses of covariance (ANCOVA) models were used to examine effects of sex and diagnosis (DX) on CT and verbal memory. For regions demonstrating significant interaction effects of sex and DX, we tested whether sex moderated cognition-thickness relationships. We used machine learning as a complementary method to explore multivariate CT differences between women and men. RESULTS Women demonstrated greater CT in many brain regions. More specifically, men showed relatively consistent CT declines in all stages, from normal control (NC) to ADD in the bilateral cingulate cortex, bilateral temporal regions, and left precuneus; women had more stable CT in these regions between NC and mild cognitive impairment (MCI) stages, but sharper declines from MCI to ADD. Similarly, for the Rey Auditory Verbal Learning Test (RAVLT), ANCOVA analyses showed that women had significantly better immediate and delayed recall scores than men, at NC and MCI stages, but greater differences, cross-sectionally, from MCI to ADD than men. We found significant sex moderation effects between RAVLT-immediate scores and CT of right isthmus-cingulate for all subjects across DX. Partial correlation analyses revealed that increased CT of right isthmus-cingulate was associated with better verbal learning in women, driven by positron emission tomography defined amyloid positive (Aβ+) subjects. Significant sex-moderation effects in cognition-thickness relationships were further found in the right middle-temporal, left precuneus, and left superior temporal regions in Aβ+ subjects. Using a machine learning approach, we investigated multivariate CT differences between women and men, showing an accuracy in classification of 75% for Aβ+ cognitively NC participants. CONCLUSIONS Sex differences in memory and CT can play a key role in the different vulnerability and progression of ADD in women compared to men. Machine learning indicates sex differences in CT are most relevant early in the ADD neurodegeneration.
Collapse
Affiliation(s)
- Filippo Cieri
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Xiaowei Zhuang
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA.,Interdisciplinary Neuroscience Program, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA
| | - Dietmar Cordes
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA.,University of Colorado Boulder, Boulder, CO, USA
| | - Nikki Kaplan
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Jeffery Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas (UNLV), Las Vegas, NV, USA
| | - Jessica Caldwell
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA.
| | | |
Collapse
|
26
|
Eastman J, Bahorik A, Kornblith E, Xia F, Yaffe K. Sex Differences in the Risk of Dementia in Older Veterans. J Gerontol A Biol Sci Med Sci 2022; 77:1250-1253. [PMID: 35134198 PMCID: PMC9391887 DOI: 10.1093/gerona/glac029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Studies have demonstrated women to have a higher prevalence of dementia compared to men. However, sex differences in dementia incidence are controversial with conflicting reports showing women with higher, lower, or similar incidence. Source of difference may be due to clinical setting and lack of consideration of competing risk of death. We examined dementia incidence in a sample of the national Veteran population to determine differences by sex. METHODS We examined data from the Veterans Health Administration (VHA), the largest integrated health care system in the United States. We studied 947 797 Veterans aged ≥55 years (mean age: 69.9 ± 8.4, 3% female) evaluated in the VHA from October 1, 1999 to September 30, 2019. We estimated age-adjusted incidence rates of dementia (International Classification of Diseases, 9th and 10th Edition codes) by sex, and used Fine-Gray proportional hazards models with age as time scale to examine time to diagnosis, accounting for competing risk of death. RESULTS During the follow-up (mean 8.4 years), 11.3% (n = 106 977, 11.4% men and 8.0% women) of Veterans developed dementia. Age-adjusted incidence was 12.6/1 000 person-years for men and 12.7/1 000 person-years for women. Compared to male Veterans, risk dementia was slightly higher among females (hazard ratio = 1.15; 95% confidence interval: 1.10-1.20), and on average, female Veterans developed dementia 0.2 years earlier than male Veterans. After additional adjustment for race, education, medical, and psychiatric conditions, results were similar. CONCLUSIONS Among older Veterans in a national cohort, women had a slightly increased risk for developing dementia compared to men after accounting for competing risk of death.
Collapse
Affiliation(s)
- Jennifer Eastman
- San Francisco Veterans Affairs Health Care System, San Francisco, California, USA,Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, California, USA
| | - Amber Bahorik
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, California, USA
| | - Erica Kornblith
- San Francisco Veterans Affairs Health Care System, San Francisco, California, USA,Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, California, USA
| | - Feng Xia
- Northern California Institute for Research and Education, San Francisco, California, USA
| | - Kristine Yaffe
- Address correspondence to: Kristine Yaffe, MD, University of California, San Francisco, San Francisco Veterans Affairs Health Care System, 4150 Clement Street, San Francisco, CA 94121, USA. E-mail:
| |
Collapse
|
27
|
Bergamino M, Keeling EG, Baxter LC, Sisco NJ, Walsh RR, Stokes AM. Sex Differences in Alzheimer's Disease Revealed by Free-Water Diffusion Tensor Imaging and Voxel-Based Morphometry. J Alzheimers Dis 2022; 85:395-414. [PMID: 34842185 PMCID: PMC9015709 DOI: 10.3233/jad-210406] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Imaging biomarkers are increasingly used in Alzheimer's disease (AD), and the identification of sex differences using neuroimaging may provide insight into disease heterogeneity, progression, and therapeutic targets. OBJECTIVE The purpose of this study was to investigate differences in grey matter (GM) volume and white matter (WM) microstructural disorganization between males and females with AD using voxel-based morphometry (VBM) and free-water-corrected diffusion tensor imaging (FW-DTI). METHODS Data were downloaded from the OASIS-3 database, including 158 healthy control (HC; 86 females) and 46 mild AD subjects (24 females). VBM and FW-DTI metrics (fractional anisotropy (FA), axial and radial diffusivities (AxD and RD, respectively), and FW index) were compared using effect size for the main effects of group, sex, and their interaction. RESULTS Significant group and sex differences were observed, with no significant interaction. Post-hoc comparisons showed that AD is associated with reduced GM volume, reduced FW-FA, and higher FW-RD/FW-index, consistent with neurodegeneration. Females in both groups exhibited higher GM volume than males, while FW-DTI metrics showed sex differences only in the AD group. Lower FW, lower FW-FA and higher FW-RD were observed in females relative to males in the AD group. CONCLUSION The combination of VBM and DTI may reveal complementary sex-specific changes in GM and WM associated with AD and aging. Sex differences in GM volume were observed for both groups, while FW-DTI metrics only showed significant sex differences in the AD group, suggesting that WM tract disorganization may play a differential role in AD pathophysiology between females and males.
Collapse
Affiliation(s)
| | - Elizabeth G. Keeling
- Neuroimaging Research, Barrow Neurological Institute,School of Life Sciences, Arizona State University
| | | | | | - Ryan R. Walsh
- Muhammad Ali Parkinson Center at Barrow Neurological
Institute
| | | |
Collapse
|
28
|
Salminen LE, Tubi MA, Bright J, Thomopoulos SI, Wieand A, Thompson PM. Sex is a defining feature of neuroimaging phenotypes in major brain disorders. Hum Brain Mapp 2022; 43:500-542. [PMID: 33949018 PMCID: PMC8805690 DOI: 10.1002/hbm.25438] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 12/12/2022] Open
Abstract
Sex is a biological variable that contributes to individual variability in brain structure and behavior. Neuroimaging studies of population-based samples have identified normative differences in brain structure between males and females, many of which are exacerbated in psychiatric and neurological conditions. Still, sex differences in MRI outcomes are understudied, particularly in clinical samples with known sex differences in disease risk, prevalence, and expression of clinical symptoms. Here we review the existing literature on sex differences in adult brain structure in normative samples and in 14 distinct psychiatric and neurological disorders. We discuss commonalities and sources of variance in study designs, analysis procedures, disease subtype effects, and the impact of these factors on MRI interpretation. Lastly, we identify key problems in the neuroimaging literature on sex differences and offer potential recommendations to address current barriers and optimize rigor and reproducibility. In particular, we emphasize the importance of large-scale neuroimaging initiatives such as the Enhancing NeuroImaging Genetics through Meta-Analyses consortium, the UK Biobank, Human Connectome Project, and others to provide unprecedented power to evaluate sex-specific phenotypes in major brain diseases.
Collapse
Affiliation(s)
- Lauren E. Salminen
- Imaging Genetics CenterMark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USCMarina del ReyCaliforniaUSA
| | - Meral A. Tubi
- Imaging Genetics CenterMark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USCMarina del ReyCaliforniaUSA
| | - Joanna Bright
- Imaging Genetics CenterMark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USCMarina del ReyCaliforniaUSA
| | - Sophia I. Thomopoulos
- Imaging Genetics CenterMark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USCMarina del ReyCaliforniaUSA
| | - Alyssa Wieand
- Imaging Genetics CenterMark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USCMarina del ReyCaliforniaUSA
| | - Paul M. Thompson
- Imaging Genetics CenterMark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USCMarina del ReyCaliforniaUSA
| |
Collapse
|
29
|
Jayaraman A, Htike TT, James R, Picon C, Reynolds R. TNF-mediated neuroinflammation is linked to neuronal necroptosis in Alzheimer's disease hippocampus. Acta Neuropathol Commun 2021; 9:159. [PMID: 34625123 PMCID: PMC8501605 DOI: 10.1186/s40478-021-01264-w] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 09/17/2021] [Indexed: 12/12/2022] Open
Abstract
The pathogenetic mechanisms underlying neuronal death and dysfunction in Alzheimer’s disease (AD) remain unclear. However, chronic neuroinflammation has been implicated in stimulating or exacerbating neuronal damage. The tumor necrosis factor (TNF) superfamily of cytokines are involved in many systemic chronic inflammatory and degenerative conditions and are amongst the key mediators of neuroinflammation. TNF binds to the TNFR1 and TNFR2 receptors to activate diverse cellular responses that can be either neuroprotective or neurodegenerative. In particular, TNF can induce programmed necrosis or necroptosis in an inflammatory environment. Although activation of necroptosis has recently been demonstrated in the AD brain, its significance in AD neuron loss and the role of TNF signaling is unclear. We demonstrate an increase in expression of multiple proteins in the TNF/TNF receptor-1-mediated necroptosis pathway in the AD post-mortem brain, as indicated by the phosphorylation of RIPK3 and MLKL, predominantly observed in the CA1 pyramidal neurons. The density of phosphoRIPK3 + and phosphoMLKL + neurons correlated inversely with total neuron density and showed significant sexual dimorphism within the AD cohort. In addition, apoptotic signaling was not significantly activated in the AD brain compared to the control brain. Exposure of human iPSC-derived glutamatergic neurons to TNF increased necroptotic cell death when apoptosis was inhibited, which was significantly reversed by small molecule inhibitors of RIPK1, RIPK3, and MLKL. In the post-mortem AD brain and in human iPSC neurons, in response to TNF, we show evidence of altered expression of proteins of the ESCRT III complex, which has been recently suggested as an antagonist of necroptosis and a possible mechanism by which cells can survive after necroptosis has been triggered. Taken together, our results suggest that neuronal loss in AD is due to TNF-mediated necroptosis rather than apoptosis, which is amenable to therapeutic intervention at several points in the signaling pathway.
Collapse
|
30
|
Luckey AM, Robertson IH, Lawlor B, Mohan A, Vanneste S. Sex Differences in Locus Coeruleus: A Heuristic Approach That May Explain the Increased Risk of Alzheimer's Disease in Females. J Alzheimers Dis 2021; 83:505-522. [PMID: 34334399 DOI: 10.3233/jad-210404] [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: 12/16/2022]
Abstract
This article aims to reevaluate our approach to female vulnerability to Alzheimer's disease (AD) and put forth a new hypothesis considering how sex differences in the locus coeruleus-noradrenaline (LC-NA) structure and function could account for why females are more likely to develop AD. We specifically focus our attention on locus coeruleus (LC) morphology, the paucity of estrogens, neuroinflammation, blood-brain barrier permeability, apolipoprotein ɛ4 polymorphism (APOEɛ4), and cognitive reserve. The role of the LC-NA system and sex differences are two of the most rapidly emerging topics in AD research. Current literature either investigates the LC due to it being one of the first brain areas to develop AD pathology or acknowledges the neuroprotective effects of estrogens and how the loss of these female hormones have the capacity to contribute to the sex differences seen in AD; however, existing research has neglected to concurrently examine these two rationales and therefore leaving our hypothesis undetermined. Collectively, this article should assist in alleviating current challenges surrounding female AD by providing thought-provoking connections into the interrelationship between the disruption of the female LC-NA system, the decline of estrogens, and AD vulnerability. It is therefore likely that treatment for this heterogeneous disease may need to be distinctly developed for females and males separately, and may require a precision medicine approach.
Collapse
Affiliation(s)
- Alison M Luckey
- Lab for Clinical & Integrative Neuroscience, School of Psychology, Trinity College Dublin, Dublin, Ireland
| | - Ian H Robertson
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| | - Brian Lawlor
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| | - Anusha Mohan
- Lab for Clinical & Integrative Neuroscience, School of Psychology, Trinity College Dublin, Dublin, Ireland
| | - Sven Vanneste
- Lab for Clinical & Integrative Neuroscience, School of Psychology, Trinity College Dublin, Dublin, Ireland.,Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland.,Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
31
|
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
|
32
|
Szoeke C, Downie SJ, Parker AF, Phillips S. Sex hormones, vascular factors and cognition. Front Neuroendocrinol 2021; 62:100927. [PMID: 34119528 DOI: 10.1016/j.yfrne.2021.100927] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 05/27/2021] [Accepted: 06/06/2021] [Indexed: 11/25/2022]
Abstract
After more than a century of research, we have failed to develop a pharmacological prevention or cure for dementia. There are strong indicators that sex hormones influence cognition. In this paper we discuss the role of these hormones at the intersection between vascular disease and dementia, in light of the mounting literature covering the shared risk factors, pathological features alongside the timeline of hormonal change with the evolution of vascular and neurodegenerative disease. Interactive risk factors and the role of inflammation over the duration of disease evolution are highlighted. Our summary tables assessing the impact of estrogen-based hormone therapy on cognition over the past 45 years illustrate the effort expended to determine the ideal age for intervention and the type, dose, administration, and duration of therapy that might improve or protect cognition as well as alleviate menopausal symptoms. As the prevalence of dementia is rising and is higher in women, it is crucial we advance our knowledge from the "inconclusive" position statement on menopausal hormone therapy of the US Preventive Services Task Force.
Collapse
Affiliation(s)
- C Szoeke
- Healthy Ageing Program, Centre for Medical Research (Royal Melbourne Hospital), Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia.
| | - S J Downie
- Healthy Ageing Program, Centre for Medical Research (Royal Melbourne Hospital), Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - A F Parker
- Department of Psychology, University of Victoria, Victoria, British Columbia, Canada
| | - S Phillips
- Healthy Ageing Program, Centre for Medical Research (Royal Melbourne Hospital), Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
33
|
Iversen WL, Cowan RL, Atalla S, Englehart SS, Gure TR, Moss KO, Ryan CM, Scharre DW, Wright KD, Monroe TB. Treating the most vulnerable: A discursive review of experimental pain in Alzheimer's disease. Nurs Open 2021; 9:942-949. [PMID: 34165251 PMCID: PMC8859087 DOI: 10.1002/nop2.922] [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: 11/06/2020] [Revised: 03/18/2021] [Accepted: 03/29/2021] [Indexed: 11/10/2022] Open
Abstract
AIM The purpose of this manuscript is to summarize research on how experimental pain is experienced by adults with Alzheimer's disease (AD) and to translate results into implications for nurses. DESIGN This discursive review synthesizes the results of three previous research studies exploring experimental pain in adults with AD. METHODS Using a series of fictional clinical vignettes, the authors discuss how the results from three previous papers using acute experimental pain can potentially be translated into clinical practice. The authors also introduce the reader to the concept of research-related psychophysics using introductory definitions and concepts with the impetus to encourage other nurses to consider this research methodology. RESULTS Pain characteristics in AD that differ from cognitively intact controls must be explored to properly address pain in this population. Nurses are well positioned to address these issues in order to provide a high quality of care to adults with AD.
Collapse
Affiliation(s)
- Wm Larkin Iversen
- The Ohio State University College of Nursing, The Ohio State University, Columbus, OH, USA
| | - Ronald L Cowan
- Department of Psychiatry, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Sebastian Atalla
- The Ohio State University College of Nursing, The Ohio State University, Columbus, OH, USA
| | - Sydney S Englehart
- The Ohio State University College of Nursing, The Ohio State University, Columbus, OH, USA
| | - Tanya R Gure
- Division of General Internal Medicine and Geriatrics, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Karen O Moss
- The Ohio State University College of Nursing, The Ohio State University, Columbus, OH, USA
| | - Claire M Ryan
- Vanderbilt Department of Psychiatry and Behavioral Sciences, Nashville, TN, USA
| | - Douglas W Scharre
- Department of Neurology, Division of Cognitive Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Kathy D Wright
- The Ohio State University College of Nursing, The Ohio State University, Columbus, OH, USA
| | - Todd B Monroe
- The Ohio State University College of Nursing, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
34
|
Wang YTT, Pascoal TA, Therriault J, Kang MS, Benedet AL, Savard M, Tissot C, Lussier FZ, Arias JF, Mathotaarachchi S, Rajah MN, Gauthier S, Rosa-Neto P. Interactive rather than independent effect of APOE and sex potentiates tau deposition in women. Brain Commun 2021; 3:fcab126. [PMID: 34189460 PMCID: PMC8226193 DOI: 10.1093/braincomms/fcab126] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/07/2021] [Accepted: 06/04/2021] [Indexed: 11/14/2022] Open
Abstract
The apolipoprotein E gene (APOE) is the most important genetic risk factor for sporadic Alzheimer disease, with the ε4 allele being associated with increased cerebral amyloid-β and tau pathologies. Although APOE has been suggested to have a stronger effect in women as compared to men, there is a lack of comprehensive assessment on how the interactive effect of APOE and sex modulates regional vulnerability to tau accumulation. We previously have shown the regional vulnerability to the interactive effect of tau and APOE, yet the sex difference was not specifically addressed. In this study, we leveraged PET imaging data from the Translational Biomarkers in Aging and Dementia cohort at McGill University Research Centre for Studies in Aging to elucidate the APOE-by-sex interactive effect on tau burden. We hypothesized sex-dependent regional vulnerability to tau deposition. PET radiopharmaceuticals [18F]AZD4694 and [18F]MK6240 were used to assess amyloid-β and tau level respectively in 277 subjects from the Translational Biomarkers in Aging and Dementia cohort. We found that the interaction between APOE and sex, rather than their independent main effects, was associated with abnormal tau accumulation in medial temporal regions. Specifically, we found that female APOEε4 carriers showed significantly higher tau burden in early tau deposition regions including the hippocampus, entorhinal and parahippocampal cortices, after accounting for age, educational attainment, clinical diagnosis and neocortical amyloid load. We replicated these findings in 221 subjects from the Alzheimer's Disease Neuroimaging Initiative cohort, in which a different tau-PET radioligand, [18F]flortaucipir, was used to assess tau burden. In conclusion, this study provides evidence from two cohort studies that interactive rather than independent effect of APOE and sex potentiates early tau deposition in women. Our results have important implications for clinical trials and practice, which should take into consideration both APOEε4 carriage status and sex for identifying individuals with the highest probability of developing tau accumulation and clinical progression.
Collapse
Affiliation(s)
- Yi-Ting T Wang
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC, Canada
- Douglas Research Centre, Montréal, QC, Canada
| | - Tharick A Pascoal
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Joseph Therriault
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC, Canada
- Douglas Research Centre, Montréal, QC, Canada
| | - Min Su Kang
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC, Canada
- Douglas Research Centre, Montréal, QC, Canada
| | - Andréa L Benedet
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC, Canada
| | - Melissa Savard
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC, Canada
| | - Cécile Tissot
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC, Canada
- Douglas Research Centre, Montréal, QC, Canada
| | - Firoza Z Lussier
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC, Canada
- Douglas Research Centre, Montréal, QC, Canada
| | - Jaime Fernandez Arias
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC, Canada
- Douglas Research Centre, Montréal, QC, Canada
| | - Sulantha Mathotaarachchi
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC, Canada
| | - Maria Natasha Rajah
- Douglas Research Centre, Montréal, QC, Canada
- Department of Psychiatry, McGill University, Montréal, QC, Canada
| | - Serge Gauthier
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC, Canada
- Douglas Research Centre, Montréal, QC, Canada
- Department of Psychiatry, McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, The McGill University Research Centre for Studies in Aging, McGill University, Montréal, QC, Canada
- Douglas Research Centre, Montréal, QC, Canada
- Department of Psychiatry, McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | | |
Collapse
|
35
|
Falcon C, Grau-Rivera O, Suárez-Calvet M, Bosch B, Sánchez-Valle R, Arenaza-Urquijo EM, González-de-Echavarri JM, Gispert JD, Rami L, Molinuevo JL. Sex Differences of Longitudinal Brain Changes in Cognitively Unimpaired Adults. J Alzheimers Dis 2021; 76:1413-1422. [PMID: 32651319 DOI: 10.3233/jad-200293] [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: 12/29/2022]
Abstract
BACKGROUND There is increasing evidence that AD progression differs by sex. OBJECTIVE The aim of this work was to determine sex differences in the association of baseline levels of cerebrospinal fluid (CSF) biomarkers (Aβ42, p-tau, YKL-40, sTREM2) with longitudinal brain changes in cognitively unimpaired (CU) older adults. METHODS This pilot study included 36 CU subjects (age 66.5±5.5, 12 male) scanned twice, two years apart. Using a voxel-wise analysis, we determined the sex differences in the association maps between CSF biomarkers and atrophy rates. RESULTS We did not find differences related to Aβ42. We found a greater impact of the rest of CSF biomarkers in areas of the Papez circuit in women versus men. Men showed greater involvement in lateral parietal and paracentral areas. DISCUSSION Results suggest an early differential progression of brain atrophy between sexes. Further research will elucidate whether the mechanisms responsible for sex-specific atrophy patterns are biological and/or environmental.
Collapse
Affiliation(s)
- Carles Falcon
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain.,Centro de Investigación Biomédicaen Red Bioingeniería, Biomateriales y Nanomedicina (CIBERBBN), Madrid, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Oriol Grau-Rivera
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,CIBERFragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain.,Servei de Neurologia, Hospital del Mar, Barcelona, Spain
| | - Marc Suárez-Calvet
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,CIBERFragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain.,Servei de Neurologia, Hospital del Mar, Barcelona, Spain
| | - Beatriz Bosch
- Alzheimer's disease and other cognitive disorders Unit, Neurology Service, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Raquel Sánchez-Valle
- Alzheimer's disease and other cognitive disorders Unit, Neurology Service, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Eider M Arenaza-Urquijo
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,CIBERFragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - José María González-de-Echavarri
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Juan Domingo Gispert
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain.,Centro de Investigación Biomédicaen Red Bioingeniería, Biomateriales y Nanomedicina (CIBERBBN), Madrid, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| | - Lorena Rami
- Alzheimer's disease and other cognitive disorders Unit, Neurology Service, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - José Luis Molinuevo
- Barcelonaβeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,CIBERFragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain.,Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
36
|
Zhu W, Huang H, Yang S, Luo X, Zhu W, Xu S, Meng Q, Zuo C, Liu Y, Wang W. Cortical and Subcortical Grey Matter Abnormalities in White Matter Hyperintensities and Subsequent Cognitive Impairment. Neurosci Bull 2021; 37:789-803. [PMID: 33826095 PMCID: PMC8192646 DOI: 10.1007/s12264-021-00657-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/28/2020] [Indexed: 01/18/2023] Open
Abstract
Grey matter (GM) alterations may contribute to cognitive decline in individuals with white matter hyperintensities (WMH) but no consensus has yet emerged. Here, we investigated cortical thickness and grey matter volume in 23 WMH patients with mild cognitive impairment (WMH-MCI), 43 WMH patients without cognitive impairment, and 55 healthy controls. Both WMH groups showed GM atrophy in the bilateral thalamus, fronto-insular cortices, and several parietal-temporal regions, and the WMH-MCI group showed more extensive and severe GM atrophy. The GM atrophy in the thalamus and fronto-insular cortices was associated with cognitive decline in the WMH-MCI patients and may mediate the relationship between WMH and cognition in WMH patients. Furthermore, the main results were well replicated in an independent dataset from the Alzheimer's Disease Neuroimaging Initiative database and in other control analyses. These comprehensive results provide robust evidence of specific GM alterations underlying WMH and subsequent cognitive impairment.
Collapse
Affiliation(s)
- Wenhao Zhu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hao Huang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shiqi Yang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiang Luo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wenzhen Zhu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shabei Xu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qi Meng
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chengchao Zuo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yong Liu
- Brainnetome Center and National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, 100190, China.
- School of Artificial Intelligence, Beijing University of Posts and Telecommunications, Beijing, 100876, China.
- University of the Chinese Academy of Sciences, Beijing, 100049, China.
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
37
|
Shao W, He X, Li X, Tao W, Zhang J, Zhang S, Wang L, Qiao Y, Wang Y, Zhang Z, Peng D. Disrupted White Matter Networks from Subjective Memory Impairment to Amnestic Mild Cognitive Impairment. Curr Alzheimer Res 2021; 18:35-44. [PMID: 33761859 DOI: 10.2174/1567205018666210324115817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 12/13/2020] [Accepted: 03/08/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND OBJECTIVE Subjective memory impairment (SMI) is a preclinical stage prior to amnestic mild cognitive impairment (aMCI) along with the Alzheimer's disease (AD) continuum. We hypothesized that SMI patients had white matter (WM) network disruptions similar to those in aMCI patients. METHODS We used diffusion-tensor magnetic resonance imaging and graph theory to construct, analyze, and compare the WM networks among 20 normal controls (NC), 20 SMI patients, and 20 aMCI patients. RESULTS Compared with the NC group, the SMI group had significantly decreased global and local efficiency and an increased shortest path length. Moreover, similar to the aMCI group, the SMI group had lower nodal efficiency in regions located in the frontal and parietal lobes, limbic systems, and caudate nucleus compared to that of the NC group. CONCLUSION Similar to aMCI patient, SMI patients exhibited WM network disruptions, and detection of these disruptions could facilitate the early detection of SMI.
Collapse
Affiliation(s)
- Wen Shao
- Department of Neurology, China-Japan Friendship Hospital, Beijing 100029,China
| | - Xuwen He
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875,China
| | - Xin Li
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875,China
| | - Wuhai Tao
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875,China
| | - Junying Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875,China
| | - Shujuan Zhang
- Department of Neurology, China-Japan Friendship Hospital, Beijing 100029,China
| | - Lei Wang
- Department of Neurology, China-Japan Friendship Hospital, Beijing 100029,China
| | - Yanan Qiao
- Department of Neurology, China-Japan Friendship Hospital, Beijing 100029,China
| | - Yu Wang
- Department of Neurology, China-Japan Friendship Hospital, Beijing 100029,China
| | - Zhanjun Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875,China
| | - Dantao Peng
- Department of Neurology, China-Japan Friendship Hospital, Beijing 100029,China
| |
Collapse
|
38
|
Early Onset of Sex-Dependent Mitochondrial Deficits in the Cortex of 3xTg Alzheimer's Mice. Cells 2020; 9:cells9061541. [PMID: 32599904 PMCID: PMC7349170 DOI: 10.3390/cells9061541] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/09/2020] [Accepted: 06/18/2020] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease (AD) is a major public health concern worldwide. Advanced age and female sex are two of the most prominent risk factors for AD. AD is characterized by progressive neuronal loss, especially in the cortex and hippocampus, and mitochondrial dysfunction has been proposed to be an early event in the onset and progression of the disease. Our results showed early perturbations in mitochondrial function in 3xTg mouse brain, with the cortex being more susceptible to mitochondrial changes than the hippocampus. In the cortex of 3xTg females, decreased coupled and uncoupled respiration were evident early (at 2 months of age), while in males it appeared later at 6 months of age. We observed increased coupled respiration in the hippocampus of 2-month-old 3xTg females, but no changes were detected later in life. Changes in mitochondrial dynamics were indicated by decreased mitofusin (Mfn2) and increased dynamin related protein 1 (Drp1) (only in females) in the hippocampus and cortex of 3xTg mice. Our findings highlight the importance of controlling and accounting for sex, brain region, and age in studies examining brain bioenergetics using this common AD model in order to more accurately evaluate potential therapies and improve the sex-specific translatability of preclinical findings.
Collapse
|
39
|
Montandon ML, Herrmann FR, Garibotto V, Rodriguez C, Haller S, Giannakopoulos P. Determinants of mesial temporal lobe volume loss in older individuals with preserved cognition: a longitudinal PET amyloid study. Neurobiol Aging 2019; 87:108-114. [PMID: 32057528 DOI: 10.1016/j.neurobiolaging.2019.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 12/03/2019] [Accepted: 12/05/2019] [Indexed: 12/11/2022]
Abstract
Mesial temporal lobe (MTL) is prominently affected in normal aging and associated with neurodegeneration in AD. Whether or not MTL atrophy is dependent on increasing amyloid load before the emergence of cognitive deficits is still disputed. We performed a 4.5-year longitudinal study in 75 older community dwellers (48 women, mean age: 79.3 years) including magnetic resonance imaging at baseline and follow-up, positron emission tomography amyloid during follow-up, neuropsychological assessment at 18 and 55 months, and APOE genotyping. Linear regression models were used to identify predictors of the MTL volume loss. Amyloid load was negatively associated with bilateral MTL volume at baseline explaining almost 10.5% of its variability. In multivariate models including time of follow-up and demographic variables (older age, male gender), this percentage exceeded 35%. The APOE4 allele independently contributed another 6%. Cognitive changes had a modest but still significant negative association with MTL volume loss. Our data support a multifactorial model including amyloid deposition, older age, male gender, APOE4 allele, and slight decline of cognitive abilities as independent predictors of MTL volume loss in brain aging.
Collapse
Affiliation(s)
- Marie-Louise Montandon
- Department of Rehabilitation and Geriatrics, Geneva University Hospitals and University of Geneva, Thônex, Switzerland; Department of Psychiatry, University of Geneva, Thônex, Switzerland
| | - François R Herrmann
- Department of Rehabilitation and Geriatrics, Geneva University Hospitals and University of Geneva, Thônex, Switzerland.
| | - Valentina Garibotto
- Division of Nuclear Medicine and Molecular Imaging, Diagnostic Department, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Cristelle Rodriguez
- Department of Psychiatry, University of Geneva, Thônex, Switzerland; Medical Direction, University of Geneva Hospitals, Geneva, Switzerland
| | - Sven Haller
- CIRD - Centre d'Imagerie Rive Droite, Geneva, Switzerland; Department of Surgical Sciences, Radiology, Uppsala University, Uppsala, Sweden; Department of Neuroradiology, Faculty of Medicine of the University of Geneva, Geneva, Switzerland
| | - Panteleimon Giannakopoulos
- Department of Psychiatry, University of Geneva, Thônex, Switzerland; Medical Direction, University of Geneva Hospitals, Geneva, Switzerland
| |
Collapse
|
40
|
Sex Differences in Neuropsychological Test Performance in Alzheimer's Disease and the Influence of the ApoE Genotype. Alzheimer Dis Assoc Disord 2019; 32:145-149. [PMID: 29189302 DOI: 10.1097/wad.0000000000000229] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Only few studies exist reporting sex differences in the Alzheimer disease (AD) patients regarding cognitive profile, brain damage, and risk factors. The present study investigated the influence of sex in combination with the Alzheimer risk allele, ε4-allele of apolipoprotein E, on cognitive performance. MATERIALS AND METHODS We examined the effect of sex and ApoE genotype on a range of neuropsychological markers from the German version of the Consortium to Establish a Registry in Alzheimer's Disease Neuropsychological Battery in a monocentric study of 399 AD patients. RESULTS Male patients had significantly more years of school and occupational education compared with women. Male AD patients outperformed female patients in tasks of object naming, constructional praxis, and constructional praxis recall. There was no statistically significant interaction effect between sex and ε4-allele of apolipoprotein E for any of the examined variables. CONCLUSIONS The superiority of healthy men compared with women in tasks of object naming, constructional praxis, and visual memory seems to remain stable when people develop AD, indicating larger cognitive reserves in men. In contrast, findings that cognitively healthy women outperform men in tests of verbal memory and verbal fluency are not stable in AD. Further studies are needed to gain insight in the reasons for sex differences.
Collapse
|
41
|
Marongiu R. Accelerated Ovarian Failure as a Unique Model to Study Peri-Menopause Influence on Alzheimer's Disease. Front Aging Neurosci 2019; 11:242. [PMID: 31551757 PMCID: PMC6743419 DOI: 10.3389/fnagi.2019.00242] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 08/19/2019] [Indexed: 12/11/2022] Open
Abstract
Despite decades of extensive research efforts, efficacious therapies for Alzheimer's disease (AD) are lacking. The multi-factorial nature of AD neuropathology and symptomatology has taught us that a single therapeutic approach will most likely not fit all. Women constitute ~70% of the affected AD population, and pathology and rate of symptoms progression are 2-3 times higher in women than men. Epidemiological data suggest that menopausal estrogen loss may be causative of the more severe symptoms observed in AD women, however, results from clinical trials employing estrogen replacement therapy are inconsistent. AD pathological hallmarks-amyloid β (Aβ), neurofibrillary tangles (NFTs), and chronic gliosis-are laid down during a 20-year prodromal period before clinical symptoms appear, which coincides with the menopause transition (peri-menopause) in women (~45-54-years-old). Peri-menopause is marked by widely fluctuating estrogen levels resulting in periods of irregular hormone-receptor interactions. Recent studies showed that peri-menopausal women have increased indicators of AD phenotype (brain Aβ deposition and hypometabolism), and peri-menopausal women who used hormone replacement therapy (HRT) had a reduced AD risk. This suggests that neuroendocrine changes during peri-menopause may be a trigger that increases risk of AD in women. Studies on sex differences have been performed in several AD rodent models over the years. However, it has been challenging to study the menopause influence on AD due to lack of optimal models that mimic the human process. Recently, the rodent model of accelerated ovarian failure (AOF) was developed, which uniquely recapitulates human menopause, including a transitional peri-AOF period with irregular estrogen fluctuations and a post-AOF stage with low estrogen levels. This model has proven useful in hypertension and cognition studies with wild type animals. This review article will highlight the molecular mechanisms by which peri-menopause may influence the female brain vulnerability to AD and AD risk factors, such as hypertension and apolipoprotein E (APOE) genotype. Studies on these biological mechanisms together with the use of the AOF model have the potential to shed light on key molecular pathways underlying AD pathogenesis for the development of precision medicine approaches that take sex and hormonal status into account.
Collapse
Affiliation(s)
- Roberta Marongiu
- Laboratory of Molecular Neurosurgery, Weill Cornell Medicine, Department of Neurosurgery, Cornell University, New York, NY, United States
| |
Collapse
|
42
|
Abstract
Alzheimer disease (AD) is characterized by wide heterogeneity in cognitive and behavioural syndromes, risk factors and pathophysiological mechanisms. Addressing this phenotypic variation will be crucial for the development of precise and effective therapeutics in AD. Sex-related differences in neural anatomy and function are starting to emerge, and sex might constitute an important factor for AD patient stratification and personalized treatment. Although the effects of sex on AD epidemiology are currently the subject of intense investigation, the notion of sex-specific clinicopathological AD phenotypes is largely unexplored. In this Review, we critically discuss the evidence for sex-related differences in AD symptomatology, progression, biomarkers, risk factor profiles and treatment. The cumulative evidence reviewed indicates sex-specific patterns of disease manifestation as well as sex differences in the rates of cognitive decline and brain atrophy, suggesting that sex is a crucial variable in disease heterogeneity. We discuss critical challenges and knowledge gaps in our current understanding. Elucidating sex differences in disease phenotypes will be instrumental in the development of a 'precision medicine' approach in AD, encompassing individual, multimodal, biomarker-driven and sex-sensitive strategies for prevention, detection, drug development and treatment.
Collapse
|
43
|
Toro CA, Zhang L, Cao J, Cai D. Sex differences in Alzheimer's disease: Understanding the molecular impact. Brain Res 2019; 1719:194-207. [PMID: 31129153 DOI: 10.1016/j.brainres.2019.05.031] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 04/10/2019] [Accepted: 05/22/2019] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disorder that presents with cognitive impairment and behavioral disturbance. Approximately 5.5 million people in the United States live with AD, most of whom are over the age of 65 with two-thirds being woman. There have been major advancements over the last decade or so in the understanding of AD neuropathological changes and genetic involvement. However, studies of sex impact in AD have not been adequately integrated into the investigation of disease development and progression. It becomes indispensable to acknowledge in both basic science and clinical research studies the importance of understanding sex-specific differences in AD pathophysiology and pathogenesis, which could guide future effort in the discovery of novel targets for AD. Here, we review the latest and most relevant literature on this topic, highlighting the importance of understanding sex dimorphism from a molecular perspective and its association to clinical trial design and development in AD research field.
Collapse
Affiliation(s)
- Carlos A Toro
- National Center for the Medical Consequences of Spinal Cord Injury, James J Peters VA Medical Center, Bronx, NY 10468, United States; Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States.
| | - Larry Zhang
- Research and Development, James J Peters VA Medical Center, Bronx, NY 10468, United States; Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Jiqing Cao
- Research and Development, James J Peters VA Medical Center, Bronx, NY 10468, United States; Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States.
| | - Dongming Cai
- Research and Development, James J Peters VA Medical Center, Bronx, NY 10468, United States; Neurology Section, James J Peters VA Medical Center, Bronx, NY 10468, United States; Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States.
| |
Collapse
|
44
|
Paranjpe MD, Chen X, Liu M, Paranjpe I, Leal JP, Wang R, Pomper MG, Wong DF, Benzinger TLS, Zhou Y. The effect of ApoE ε4 on longitudinal brain region-specific glucose metabolism in patients with mild cognitive impairment: a FDG-PET study. Neuroimage Clin 2019; 22:101795. [PMID: 30991617 PMCID: PMC6449776 DOI: 10.1016/j.nicl.2019.101795] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 03/12/2019] [Accepted: 03/24/2019] [Indexed: 11/02/2022]
Abstract
While the ApoE ε4 allele is a known risk factor for mild cognitive impairment (MCI) and Alzheimer's disease, brain region specific effects remain elusive. In this study, we investigate whether the ApoE ε4 allele exhibits brain region specific effects in longitudinal glucose uptake among patients with MCI from the Alzheimer's Disease Neuroimaging Initiative (ADNI). Preprocessed FDG PET images, MRIs, and demographic information were downloaded from the ADNI database. An iterative reblurred Van Cittertiteration method was used for partial volume correction (PVC) on all PET images. Structural MRIs were used for PET spatial normalization and region of interest (ROI) definition in standard space. Longitudinal changes in ROI FDG standardized uptake value ratio (SUVR) relative to cerebellum in 24 ApoE ε4 carriers and 24 age-matched ApoE ε4 non-carriers were measured for up to 84-months (median 72 months, SD = 11.2 months) and compared using a generalized linear mixed effects model controlling for gender, education, baseline age, and follow-up period. Additionally, voxelwise analysis was performed by implementing a paired t-test comparing matched baseline and 72 month FDG SUVR images in ApoE carriers and non-carriers separately. Results with PVC were compared with ones from non-PVC based analysis. After applying PVC, the superior fontal, parietal, lateral temporal, medial temporal, caudate, thalamus, and post-cingulate, and amygdala regions had greater longitudinal decreases in FDG uptake in ApoE ε4 carriers with MCI compared to non-carriers with MCI. Similar forebrain and limbic clusters were found through voxelwise analysis. Compared to the PVC based analysis, fewer significant ApoE-associated regions and clusters were found in the non-PVC based PET analysis. Our findings suggest that the ApoE ε4 genotype is associated with a longitudinal decline in glucose uptake in 8 forebrain and limbic brain regions in the context of MCI. In conclusion, this 84-months longitudinal FDG PET study demonstrates a novel ApoE ε4-associated brain-region specific glucose metabolism pattern in patients with MCI. Partial volume correction improved FDG PET quantification.
Collapse
Affiliation(s)
- Manish D Paranjpe
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Xueqi Chen
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| | - Min Liu
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States; Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| | - Ishan Paranjpe
- Icahn School of Medicine at Mount Sinai, NY, New York, United States
| | - Jeffrey P Leal
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Rongfu Wang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| | - Martin G Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Dean F Wong
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, United States
| | - Yun Zhou
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States; Department of Nuclear Medicine, Peking University First Hospital, Beijing, China; Mallinckrodt Institute of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, United States.
| |
Collapse
|
45
|
Müller-Gerards D, Weimar C, Abramowski J, Tebrügge S, Jokisch M, Dragano N, Erbel R, Jöckel KH, Moebus S, Winkler A. Subjective cognitive decline, APOE ε4, and incident mild cognitive impairment in men and women. ALZHEIMER'S & DEMENTIA: DIAGNOSIS, ASSESSMENT & DISEASE MONITORING 2019; 11:221-230. [PMID: 30891488 PMCID: PMC6404645 DOI: 10.1016/j.dadm.2019.01.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Introduction Possible joint effects of subjective cognitive decline (SCD) and apolipoprotein E (APOE) ε4 genotype on incident mild cognitive impairment (MCI) were examined for men and women separately. Methods Cognitively normal participants with and without SCD were included from the first follow-up examination of the population-based Heinz Nixdorf Recall study. Sex-stratified logistic regression models estimated main effects and interactions (additive, multiplicative) of SCD at the first follow-up (yes+/no−) and APOE ε4 (positive+/negative−) groups for MCI 5 years later. Results Odds for MCI 5 years later were higher in SCD/APOE ε4 group +/+ than the sum of groups +/− and −/+ in women, with a trend for positive interaction. Odds for incident MCI in men was highest in group +/−, with no interaction effect. Discussion Our findings indicate that APOE ε4 may play an important role in the association of SCD and incident MCI, especially considering sex. Further studies need to examine these associations with larger sample sizes.
Collapse
Affiliation(s)
- Diana Müller-Gerards
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Christian Weimar
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Jessica Abramowski
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sarah Tebrügge
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Martha Jokisch
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Nico Dragano
- Institute of Medical Sociology, Centre for Health and Society, University of Düsseldorf, Medical Faculty, University of Düsseldorf, Düsseldorf, Germany
| | - Raimund Erbel
- Institute of Medical Informatics, Biometrics and Epidemiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Karl-Heinz Jöckel
- Institute of Medical Informatics, Biometrics and Epidemiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Susanne Moebus
- Institute of Medical Informatics, Biometrics and Epidemiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Angela Winkler
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | | |
Collapse
|
46
|
Chen XY, Du YF, Chen L. Neuropeptides Exert Neuroprotective Effects in Alzheimer's Disease. Front Mol Neurosci 2019; 11:493. [PMID: 30687008 PMCID: PMC6336706 DOI: 10.3389/fnmol.2018.00493] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 12/21/2018] [Indexed: 01/03/2023] Open
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disorder characterized by cognitive deficits and neuronal loss. Deposition of beta-amyloid peptide (Aβ) causes neurotoxicity through the formation of plaques in brains of Alzheimer's disease. Numerous studies have indicated that the neuropeptides including ghrelin, neurotensin, pituitary adenylate cyclase-activating polypeptide (PACAP), neuropeptide Y, substance P and orexin are closely related to the pathophysiology of Alzheimer's disease. The levels of neuropeptides and their receptors change in Alzheimer's disease. These neuropeptides exert neuroprotective roles mainly through preventing Aβ accumulation, increasing neuronal glucose transport, increasing the production of neurotrophins, inhibiting endoplasmic reticulum stress and autophagy, modulating potassium channel activity and hippocampal long-term potentiation. Therefore, the neuropeptides may function as potential drug targets in the prevention and cure of Alzheimer's disease.
Collapse
Affiliation(s)
- Xin-Yi Chen
- Department of Physiology and Pathophysiology, Qingdao University, Qingdao, China.,Department of Neurology, Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Yi-Feng Du
- Department of Neurology, Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Lei Chen
- Department of Physiology and Pathophysiology, Qingdao University, Qingdao, China
| |
Collapse
|
47
|
Sinha N, Reagh ZM, Tustison NJ, Berg CN, Shaw A, Myers CE, Hill D, Yassa MA, Gluck MA. ABCA7 risk variant in healthy older African Americans is associated with a functionally isolated entorhinal cortex mediating deficient generalization of prior discrimination training. Hippocampus 2018; 29:527-538. [PMID: 30318785 DOI: 10.1002/hipo.23042] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 09/06/2018] [Accepted: 10/02/2018] [Indexed: 11/06/2022]
Abstract
Using high-resolution resting state functional magnetic resonance imaging (fMRI), the present study tested the hypothesis that ABCA7 genetic risk differentially affects intra-medial temporal lobe (MTL) functional connectivity between MTL subfields, versus internetwork connectivity of the MTL with the medial prefrontal cortex (mPFC), in nondemented older African Americans. Although the association of ABCA7 risk variants with Alzheimer's disease (AD) has been confirmed worldwide, its effect size on the relative odds of being diagnosed with AD is significantly higher in African Americans. However, little is known about the neural correlates of cognitive function in older African Americans and how they relate to AD risk conferred by ABCA7. In a case-control fMRI study of 36 healthy African Americans, we observed ABCA7 related impairments in behavioral generalization that was mediated by dissociation in entorhinal cortex (EC) resting state functional connectivity. Specifically, ABCA7 risk variant was associated with EC-hippocampus hyper-synchronization and EC-mPFC hypo-synchronization. Carriers of the risk genotype also had a significantly smaller anterolateral EC, despite our finding no group differences on standardized neuropsychological tests. Our findings suggest a model where impaired cortical connectivity leads to a more functionally isolated EC at rest, which translates into aberrant EC-hippocampus hyper-synchronization resulting in generalization deficits. While we cannot identify the exact mechanism underlying the observed alterations in EC structure and network function, considering the relevance of Aβ in ABCA7 related AD pathogenesis, the results of our study may reflect the synergistic reinforcement between amyloid and tau pathology in the EC, which significantly increases tau-induced neuronal loss and accelerates synaptic alterations. Finally, our results add to a growing literature suggesting that generalization of learning may be a useful tool for assessing the mild cognitive deficits seen in the earliest phases of prodromal AD, even before the more commonly reported deficits in episodic memory arise.
Collapse
Affiliation(s)
- Neha Sinha
- Center for Molecular and Behavioral Neuroscience, Rutgers University-Newark, Newark, New Jersey
| | - Zachariah M Reagh
- Department of Neurology, Center for Neuroscience, University of California, Davis, California
| | - Nicholas J Tustison
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia.,Center for the Neurobiology of Learning and Memory, Department of Neurobiology and Behavior, Psychiatry and Neurology, University of California, Irvine, California
| | - Chelsie N Berg
- Center for Molecular and Behavioral Neuroscience, Rutgers University-Newark, Newark, New Jersey
| | - Ashlee Shaw
- Center for Molecular and Behavioral Neuroscience, Rutgers University-Newark, Newark, New Jersey
| | - Catherine E Myers
- Neurobiology Research Laboratory VA New Jersey Health Care System East Orange, NJ.,Pharmacology Physiology and Neuroscience, Rutgers-New Jersey Medical School, Newark, New Jersey
| | - Diane Hill
- Office of University-Community Partnerships, Rutgers University-Newark, Newark, New Jersey
| | - Michael A Yassa
- Center for the Neurobiology of Learning and Memory, Department of Neurobiology and Behavior, Psychiatry and Neurology, University of California, Irvine, California
| | - Mark A Gluck
- Center for Molecular and Behavioral Neuroscience, Rutgers University-Newark, Newark, New Jersey
| |
Collapse
|
48
|
Zhen D, Xia W, Yi ZQ, Zhao PW, Zhong JG, Shi HC, Li HL, Dai ZY, Pan PL. Alterations of brain local functional connectivity in amnestic mild cognitive impairment. Transl Neurodegener 2018; 7:26. [PMID: 30443345 PMCID: PMC6220503 DOI: 10.1186/s40035-018-0134-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 10/11/2018] [Indexed: 11/10/2022] Open
Abstract
Background Resting-state functional magnetic resonance imaging studies using a regional homogeneity (ReHo) method have reported that amnestic mild cognitive impairment (aMCI) was associated with abnormalities in local functional connectivity. However, their results were not conclusive. Methods Seed-based d Mapping was used to conduct a coordinate-based meta-analysis to identify consistent ReHo alterations in aMCI. Results We identified 10 studies with 11 datasets suitable for inclusion, including 378 patients with aMCI and 435 healthy controls. This meta-analysis identified significant ReHo alterations in patients with aMCI relative to healthy controls, mainly within the default mode network (DMN) (bilateral posterior cingulate cortex [PCC], right angular gyrus, bilateral middle temporal gyri, and left parahippocampal gyrus/hippocampus), executive control network (right superior parietal lobule and dorsolateral prefrontal cortex), visual network (right lingual gyrus and left middle occipital gyrus), and sensorimotor network (right paracentral lobule/supplementary motor area, right postcentral gyrus and left posterior insula). Significant heterogeneity of ReHo alterations in the bilateral PCC, left parahippocampal gyrus/hippocampus, and right superior parietal lobule/angular gyrus was observed. Exploratory meta-regression analyses indicated that general cognitive function, gender distribution, age, and education level partially contributed to this heterogeneity. Conclusions This study provides provisional evidence that aMCI is associated with abnormal ReHo within the DMN, executive control network, visual network, and sensorimotor network. These local functional connectivity alterations suggest coexistence of functional deficits and compensation in these networks. These findings contribute to the modeling of brain functional connectomes and to a better understanding of the neural substrates of aMCI. Confounding factors merit much attention and warrant future investigations. Electronic supplementary material The online version of this article (10.1186/s40035-018-0134-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dan Zhen
- 1School of Nursing, Jiangsu Vocational College of Medicine, Yancheng, People's Republic of China
| | - Wei Xia
- 2Department of Neurology, Affiliated Yancheng Hospital, School of Medicine, Southeast University, West Xindu Road 2#, Yancheng, Jiangsu Province, 224001 People's Republic of China
| | - Zhong Quan Yi
- 2Department of Neurology, Affiliated Yancheng Hospital, School of Medicine, Southeast University, West Xindu Road 2#, Yancheng, Jiangsu Province, 224001 People's Republic of China
| | - Pan Wen Zhao
- 2Department of Neurology, Affiliated Yancheng Hospital, School of Medicine, Southeast University, West Xindu Road 2#, Yancheng, Jiangsu Province, 224001 People's Republic of China
| | - Jian Guo Zhong
- 3Department of Central Laboratory, Affiliated Yancheng Hospital, School of Medicine, Southeast University, West Xindu Road 2#, Yancheng, Jiangsu Province, 224001 People's Republic of China
| | - Hai Cun Shi
- 3Department of Central Laboratory, Affiliated Yancheng Hospital, School of Medicine, Southeast University, West Xindu Road 2#, Yancheng, Jiangsu Province, 224001 People's Republic of China
| | - Hua Liang Li
- 3Department of Central Laboratory, Affiliated Yancheng Hospital, School of Medicine, Southeast University, West Xindu Road 2#, Yancheng, Jiangsu Province, 224001 People's Republic of China
| | - Zhen Yu Dai
- 4Department of Radiology, Affiliated Yancheng Hospital, School of Medicine, Southeast University, West Xindu Road 2#, Yancheng, Jiangsu Province, 224001 People's Republic of China
| | - Ping Lei Pan
- 2Department of Neurology, Affiliated Yancheng Hospital, School of Medicine, Southeast University, West Xindu Road 2#, Yancheng, Jiangsu Province, 224001 People's Republic of China.,3Department of Central Laboratory, Affiliated Yancheng Hospital, School of Medicine, Southeast University, West Xindu Road 2#, Yancheng, Jiangsu Province, 224001 People's Republic of China
| |
Collapse
|
49
|
Nebel RA, Aggarwal NT, Barnes LL, Gallagher A, Goldstein JM, Kantarci K, Mallampalli MP, Mormino EC, Scott L, Yu WH, Maki PM, Mielke MM. Understanding the impact of sex and gender in Alzheimer's disease: A call to action. Alzheimers Dement 2018; 14:1171-1183. [PMID: 29907423 PMCID: PMC6400070 DOI: 10.1016/j.jalz.2018.04.008] [Citation(s) in RCA: 500] [Impact Index Per Article: 71.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 04/05/2018] [Accepted: 04/09/2018] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Precision medicine methodologies and approaches have advanced our understanding of the clinical presentation, development, progression, and management of Alzheimer's disease (AD) dementia. However, sex and gender have not yet been adequately integrated into many of these approaches. METHODS The Society for Women's Health Research Interdisciplinary Network on AD, comprised of an expert panel of scientists and clinicians, reviewed ongoing and published research related to sex and gender differences in AD. RESULTS The current review is a result of this Network's efforts and aims to: (1) highlight the current state-of-the-science in the AD field on sex and gender differences; (2) address knowledge gaps in assessing sex and gender differences; and (3) discuss 12 priority areas that merit further research. DISCUSSION The exclusion of sex and gender has impeded faster advancement in the detection, treatment, and care of AD across the clinical spectrum. Greater attention to these differences will improve outcomes for both sexes.
Collapse
Affiliation(s)
- Rebecca A Nebel
- Scientific Programs, Society for Women's Health Research (SWHR®), Washington, DC, USA.
| | - Neelum T Aggarwal
- Department of Neurological Sciences and the Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Lisa L Barnes
- Department of Neurological Sciences and the Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Aimee Gallagher
- Scientific Programs, Society for Women's Health Research (SWHR®), Washington, DC, USA
| | - Jill M Goldstein
- Department of Psychiatry, Harvard Medical School, and Massachusetts General Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, and Massachusetts General Hospital, Boston, MA, USA
| | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Monica P Mallampalli
- Scientific Programs, Society for Women's Health Research (SWHR®), Washington, DC, USA
| | - Elizabeth C Mormino
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Laura Scott
- Cellular and Molecular Medicine Program, Johns Hopkins University, Baltimore, MD, USA
| | - Wai Haung Yu
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Pauline M Maki
- Department of Psychology, University of Illinois at Chicago, Chicago, IL, USA; Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Michelle M Mielke
- Department of Epidemiology, Mayo Clinic, Rochester, MN, USA; Department of Neurology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
50
|
Cavedo E, Chiesa PA, Houot M, Ferretti MT, Grothe MJ, Teipel SJ, Lista S, Habert M, Potier M, Dubois B, Hampel H. Sex differences in functional and molecular neuroimaging biomarkers of Alzheimer's disease in cognitively normal older adults with subjective memory complaints. Alzheimers Dement 2018; 14:1204-1215. [DOI: 10.1016/j.jalz.2018.05.014] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 05/22/2018] [Accepted: 05/26/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Enrica Cavedo
- AXA Research Fund & Sorbonne University ChairParisFrance
- Sorbonne University, GRC n° 21Alzheimer Precision Medicine (APM)AP‐HPPitié‐Salpêtrière HospitalBoulevard de l'hôpitalParisFrance
- Brain & Spine Institute (ICM)INSERM U 1127CNRS UMR 7225ParisFrance
- Institute of Memory and Alzheimer's Disease (IM2A)Department of NeurologyPitié‐Salpêtrière HospitalAP‐HPParisFrance
- Laboratory of Alzheimer's Neuroimaging and EpidemiologyIRCCS Centro San Giovanni di Dio FatebenefratelliBresciaItaly
| | - Patrizia A. Chiesa
- AXA Research Fund & Sorbonne University ChairParisFrance
- Sorbonne University, GRC n° 21Alzheimer Precision Medicine (APM)AP‐HPPitié‐Salpêtrière HospitalBoulevard de l'hôpitalParisFrance
- Brain & Spine Institute (ICM)INSERM U 1127CNRS UMR 7225ParisFrance
- Institute of Memory and Alzheimer's Disease (IM2A)Department of NeurologyPitié‐Salpêtrière HospitalAP‐HPParisFrance
| | - Marion Houot
- Sorbonne University, GRC n° 21Alzheimer Precision Medicine (APM)AP‐HPPitié‐Salpêtrière HospitalBoulevard de l'hôpitalParisFrance
- Institute of Memory and Alzheimer's Disease (IM2A)Centre of Excellence of Neurodegenerative Disease (CoEN)ICMCIC NeurosciencesAPHP Department of NeurologyHopital Pitié‐SalpêtrièreUniversity Paris 6ParisFrance
| | - Maria Teresa Ferretti
- Institute for Regenerative Medicine University of ZurichSchlierenSwitzerland
- Neuroscience Center ZurichZurichSwitzerland
- Women's Brain ProjectSwitzerland
| | - Michel J. Grothe
- German Center for Neurodegenerative Diseases (DZNE) – Rostock/GreifswaldRostockGermany
- Department of Psychosomatic MedicineUniversity of RostockRostockGermany
| | - Stefan J. Teipel
- German Center for Neurodegenerative Diseases (DZNE) – Rostock/GreifswaldRostockGermany
- Department of Psychosomatic MedicineUniversity of RostockRostockGermany
| | - Simone Lista
- AXA Research Fund & Sorbonne University ChairParisFrance
- Sorbonne University, GRC n° 21Alzheimer Precision Medicine (APM)AP‐HPPitié‐Salpêtrière HospitalBoulevard de l'hôpitalParisFrance
- Brain & Spine Institute (ICM)INSERM U 1127CNRS UMR 7225ParisFrance
- Institute of Memory and Alzheimer's Disease (IM2A)Department of NeurologyPitié‐Salpêtrière HospitalAP‐HPParisFrance
| | - Marie‐Odile Habert
- Sorbonne UniversitésUPMC Univ Paris 06CNRSINSERMLaboratoire d'Imagerie BiomédicaleParisFrance
- Centre pour l'Acquisition et le Traitement des ImagesParisFrance
- AP‐HPHôpital Pitié‐SalpêtrièreDépartement de Médecine NucléaireParisFrance
| | - Marie‐Claude Potier
- ICM Institut du Cerveau et de la Moelle épinièreCNRS UMR7225INSERM U1127UPMCHôpital de la Pitié‐SalpêtrièreParisFrance
| | - Bruno Dubois
- Sorbonne University, GRC n° 21Alzheimer Precision Medicine (APM)AP‐HPPitié‐Salpêtrière HospitalBoulevard de l'hôpitalParisFrance
- Brain & Spine Institute (ICM)INSERM U 1127CNRS UMR 7225ParisFrance
- Institute of Memory and Alzheimer's Disease (IM2A)Department of NeurologyPitié‐Salpêtrière HospitalAP‐HPParisFrance
| | - Harald Hampel
- AXA Research Fund & Sorbonne University ChairParisFrance
- Sorbonne University, GRC n° 21Alzheimer Precision Medicine (APM)AP‐HPPitié‐Salpêtrière HospitalBoulevard de l'hôpitalParisFrance
- Brain & Spine Institute (ICM)INSERM U 1127CNRS UMR 7225ParisFrance
- Institute of Memory and Alzheimer's Disease (IM2A)Department of NeurologyPitié‐Salpêtrière HospitalAP‐HPParisFrance
| | | | | |
Collapse
|