1
|
Panagaki T, Janickova L, Petrovic D, Zuhra K, Ditrói T, Jurányi EP, Bremer O, Ascenção K, Philipp TM, Nagy P, Filipovic MR, Szabo C. Neurobehavioral dysfunction in a mouse model of Down syndrome: upregulation of cystathionine β-synthase, H 2S overproduction, altered protein persulfidation, synaptic dysfunction, endoplasmic reticulum stress, and autophagy. GeroScience 2024; 46:4275-4314. [PMID: 38558215 PMCID: PMC11336008 DOI: 10.1007/s11357-024-01146-8] [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: 02/14/2024] [Accepted: 03/21/2024] [Indexed: 04/04/2024] Open
Abstract
Down syndrome (DS) is a genetic condition where the person is born with an extra chromosome 21. DS is associated with accelerated aging; people with DS are prone to age-related neurological conditions including an early-onset Alzheimer's disease. Using the Dp(17)3Yey/ + mice, which overexpresses a portion of mouse chromosome 17, which encodes for the transsulfuration enzyme cystathionine β-synthase (CBS), we investigated the functional role of the CBS/hydrogen sulfide (H2S) pathway in the pathogenesis of neurobehavioral dysfunction in DS. The data demonstrate that CBS is higher in the brain of the DS mice than in the brain of wild-type mice, with primary localization in astrocytes. DS mice exhibited impaired recognition memory and spatial learning, loss of synaptosomal function, endoplasmic reticulum stress, and autophagy. Treatment of mice with aminooxyacetate, a prototypical CBS inhibitor, improved neurobehavioral function, reduced the degree of reactive gliosis in the DS brain, increased the ability of the synaptosomes to generate ATP, and reduced endoplasmic reticulum stress. H2S levels in the brain of DS mice were higher than in wild-type mice, but, unexpectedly, protein persulfidation was decreased. Many of the above alterations were more pronounced in the female DS mice. There was a significant dysregulation of metabolism in the brain of DS mice, which affected amino acid, carbohydrate, lipid, endocannabinoid, and nucleotide metabolites; some of these alterations were reversed by treatment of the mice with the CBS inhibitor. Thus, the CBS/H2S pathway contributes to the pathogenesis of neurological dysfunction in DS in the current animal model.
Collapse
Affiliation(s)
- Theodora Panagaki
- Chair of Pharmacology, Section of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Lucia Janickova
- Chair of Pharmacology, Section of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Dunja Petrovic
- Leibniz-Institut Für Analytische Wissenschaften-ISAS-E.V., Dortmund, Germany
| | - Karim Zuhra
- Chair of Pharmacology, Section of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Tamás Ditrói
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, Hungary
| | - Eszter P Jurányi
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, Hungary
- Doctoral School of Semmelweis University, Semmelweis University, Budapest, Hungary
| | - Olivier Bremer
- Chair of Pharmacology, Section of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Kelly Ascenção
- Chair of Pharmacology, Section of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Thilo M Philipp
- Chair of Pharmacology, Section of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Péter Nagy
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, Hungary
- Department of Anatomy and Histology, HUN-REN-UVMB Laboratory of Redox Biology Research Group, University of Veterinary Medicine, Budapest, Hungary
- Chemistry Institute, University of Debrecen, Debrecen, Hungary
| | - Milos R Filipovic
- Leibniz-Institut Für Analytische Wissenschaften-ISAS-E.V., Dortmund, Germany
| | - Csaba Szabo
- Chair of Pharmacology, Section of Science and Medicine, University of Fribourg, Fribourg, Switzerland.
| |
Collapse
|
2
|
Du Y, Yuan Z, Sui J, Calhoun VD. Common and unique brain aging patterns between females and males quantified by large-scale deep learning. Hum Brain Mapp 2024; 45:e70005. [PMID: 39225381 PMCID: PMC11369911 DOI: 10.1002/hbm.70005] [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: 02/03/2024] [Revised: 07/20/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
There has been extensive evidence that aging affects human brain function. However, there is no complete picture of what brain functional changes are mostly related to normal aging and how aging affects brain function similarly and differently between males and females. Based on resting-state brain functional connectivity (FC) of 25,582 healthy participants (13,373 females) aged 49-76 years from the UK Biobank project, we employ deep learning with explainable AI to discover primary FCs related to progressive aging and reveal similarity and difference between females and males in brain aging. Using a nested cross-validation scheme, we conduct 4200 deep learning models to classify all paired age groups on the main data for females and males separately and then extract gender-common and gender-specific aging-related FCs. Next, we validate those FCs using additional 21,000 classifiers on the independent data. Our results support that aging results in reduced brain functional interactions for both females and males, primarily relating to the positive connectivity within the same functional domain and the negative connectivity between different functional domains. Regions linked to cognitive control show the most significant age-related changes in both genders. Unique aging effects in males and females mainly involve the interaction between cognitive control and the default mode, vision, auditory, and frontoparietal domains. Results also indicate females exhibit faster brain functional changes than males. Overall, our study provides new evidence about common and unique patterns of brain aging in females and males.
Collapse
Affiliation(s)
- Yuhui Du
- School of Computer and Information TechnologyShanxi UniversityTaiyuanChina
- Tri‐Institutional Center for Translational Research in Neuroimaging and Data ScienceGeorgia State University, Georgia Institute of Technology, Emory UniversityAtlantaGeorgiaUSA
| | - Zhen Yuan
- School of Computer and Information TechnologyShanxi UniversityTaiyuanChina
| | - Jing Sui
- State Key Laboratory of Cognitive Neuroscience and LearningBeijing Normal UniversityBeijingChina
| | - Vince D. Calhoun
- Tri‐Institutional Center for Translational Research in Neuroimaging and Data ScienceGeorgia State University, Georgia Institute of Technology, Emory UniversityAtlantaGeorgiaUSA
| |
Collapse
|
3
|
Kuhn MK, Proctor EA. Microglial Drivers of Alzheimer's Disease Pathology: An Evolution of Diverse Participating States. Proteins 2024. [PMID: 39219300 DOI: 10.1002/prot.26723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 06/05/2024] [Accepted: 06/12/2024] [Indexed: 09/04/2024]
Abstract
Microglia, the resident immune-competent cells of the brain, become dysfunctional in Alzheimer's disease (AD), and their aberrant immune responses contribute to the accumulation of pathological proteins and neuronal injury. Genetic studies implicate microglia in the development of AD, prompting interest in developing immunomodulatory therapies to prevent or ameliorate disease. However, microglia take on diverse functional states in disease, playing both protective and detrimental roles in AD, which largely overlap and may shift over the disease course, complicating the identification of effective therapeutic targets. Extensive evidence gathered using transgenic mouse models supports an active role of microglia in pathology progression, though results vary and can be contradictory between different types of models and the degree of pathology at the time of study. Here, we review microglial immune signaling and responses that contribute to the accumulation and spread of pathological proteins or directly affect neuronal health. We additionally explore the use of induced pluripotent stem cell (iPSC)-derived models to study living human microglia and how they have contributed to our knowledge of AD and may begin to fill in the gaps left by mouse models. Ultimately, mouse and iPSC-derived models have their own limitations, and a comprehensive understanding of microglial dysfunction in AD will only be established by an integrated view across models and an appreciation for their complementary viewpoints and limitations.
Collapse
Affiliation(s)
- Madison K Kuhn
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Neurosurgery, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Elizabeth A Proctor
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Neurosurgery, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Engineering Science & Mechanics, The Pennsylvania State University, University Park, Pennsylvania, USA
- Penn State Neuroscience Institute, The Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
4
|
Cunha C, Rodrigues P, Voss G, Martinez-Pecino R, Delerue-Matos A. Association between formal social participation and cognitive function in middle-aged and older adults: a longitudinal study using SHARE data. NEUROPSYCHOLOGY, DEVELOPMENT, AND COGNITION. SECTION B, AGING, NEUROPSYCHOLOGY AND COGNITION 2024; 31:932-955. [PMID: 38402630 DOI: 10.1080/13825585.2024.2315769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 12/14/2023] [Indexed: 02/27/2024]
Abstract
Formal social participation significantly impacts health and well-being, potentially mitigating cognitive decline, although not consistently across all studies. Existing research often focuses solely on baseline participation levels, and age-related differences have primarily been explored among the Asian population. Therefore, this longitudinal study aims to assess the association between formal social participation and cognition across different age groups in individuals aged 50+ living in Europe and Israel, while capturing the dynamic nature of formal social participation. We use data from three waves (four, six, and eight) of the Survey of Health, Ageing, and Retirement in Europe (SHARE), comprising 85,601 respondents. Linear mixed-effects models were applied. The results show that participation in formal social activities mitigates cognitive decline in middle-aged and older adults, especially among those aged 70 to 79 and 80+. These findings support the need for social policies promoting formal social activities, for lasting cognitive health benefits.
Collapse
Affiliation(s)
- Cláudia Cunha
- Communication and Society Research Centre, Institute of Social Sciences, University of Minho, Braga, Portugal
| | - Paula Rodrigues
- Communication and Society Research Centre, Institute of Social Sciences, University of Minho, Braga, Portugal
| | - Gina Voss
- Faculty of Medicine, University of Porto, Porto, Portugal
| | | | - Alice Delerue-Matos
- Communication and Society Research Centre, Institute of Social Sciences, University of Minho, Braga, Portugal
- Department of Sociology, Institute of Social Sciences, University of Minho, Braga, Portugal
| |
Collapse
|
5
|
Song JY, Jia Y, Han H, Yang XH, Zhang J, Zhang Q, Wang SS, Wang CY, Chen L, Zhang M. Increased expression of SLC25A18 is associated with Alzheimer's disease and is involved in Aβ42-induced mitochondrial dysfunction and apoptosis in neuronal cells. Mitochondrion 2024; 78:101918. [PMID: 38871013 DOI: 10.1016/j.mito.2024.101918] [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: 11/30/2023] [Revised: 06/03/2024] [Accepted: 06/11/2024] [Indexed: 06/15/2024]
Abstract
Alzheimer's disease (AD) is currently one of the most serious public health concerns in the world. However, the best approach to treat AD has yet to be discovered, implying that we must continue to work hard to find new AD target genes. In this study, we further analysed Gene Expression Omnibus (GEO) data and discovered that the expression of the Mitochondria glutamate carrier SLC25A18 is associated with AD by screening the differentially expressed genes in different regions of the brains of Alzheimer's disease patients. To verify the expression of SLC25A18 during Alzheimer's disease development, we analysed animal models (5×FAD transgenic AD animal model, chemically induced AD animal model, natural ageing animal model), and the results showed that the expression of SLC25A18 was increased in animal models of AD. Further investigation of the different regions found that SLC25A18 expression was elevated in the EC, TeA, and CA3, and expressed in neurons. Next, We found that Aβ42 treatment elevated SLC25A18 expression in Neuro 2A cells. Reducing SLC25A18 expression attenuated mitochondrial dysfunction and neuronal apoptosis caused by Aβ42. Overexpression of SLC25A18 increased ATP and intracellular superoxide anions but decreased mitochondrial membrane potential. The results indicate that SLC25A18 affects mitochondrial function and neuronal apoptosis, and is related to AD, which makes it a potential target for treating brain dysfunction.
Collapse
Affiliation(s)
- Jia-Yi Song
- Department of Pharmacology, Basic College of Medicine, Jilin University, Changchun, Jilin Province, China; Department of General Practice, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Yong Jia
- School of nursing, Jilin University, Changchun, Jilin Province, China
| | - Hao Han
- Department of Pharmacology, Basic College of Medicine, Jilin University, Changchun, Jilin Province, China
| | - Xue-Han Yang
- Department of Pharmacology, Basic College of Medicine, Jilin University, Changchun, Jilin Province, China
| | - Jing Zhang
- Department of Pharmacology, Basic College of Medicine, Jilin University, Changchun, Jilin Province, China
| | - Qiang Zhang
- Department of Pharmacology, Basic College of Medicine, Jilin University, Changchun, Jilin Province, China
| | - Su-Shan Wang
- Department of Pharmacology, Basic College of Medicine, Jilin University, Changchun, Jilin Province, China
| | - Chun-Yan Wang
- Department of General Practice, The First Hospital of Jilin University, Changchun, Jilin Province, China.
| | - Li Chen
- Department of Pharmacology, Basic College of Medicine, Jilin University, Changchun, Jilin Province, China.
| | - Ming Zhang
- Department of Pharmacology, Basic College of Medicine, Jilin University, Changchun, Jilin Province, China.
| |
Collapse
|
6
|
Nagy B, Protzner AB, Czigler B, Gaál ZA. Resting-state neural dynamics changes in older adults with post-COVID syndrome and the modulatory effect of cognitive training and sex. GeroScience 2024:10.1007/s11357-024-01324-8. [PMID: 39210163 DOI: 10.1007/s11357-024-01324-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
Post-COVID syndrome manifests with numerous neurological and cognitive symptoms, the precise origins of which are still not fully understood. As females and older adults are more susceptible to developing this condition, our study aimed to investigate how post-COVID syndrome alters intrinsic brain dynamics in older adults and whether biological sex and cognitive training might modulate these effects, with a specific focus on older females. The participants, aged between 60 and 75 years, were divided into three experimental groups: healthy old female, post-COVID old female and post-COVID old male. They underwent an adaptive task-switching training protocol. We analysed multiscale entropy and spectral power density of resting-state EEG data collected before and after the training to assess neural signal complexity and oscillatory power, respectively. We found no difference between post-COVID females and males before training, indicating that post-COVID similarly affected both sexes. However, cognitive training was effective only in post-COVID females and not in males, by modulating local neural processing capacity. This improvement was further evidenced by comparing healthy and post-COVID females, wherein the latter group showed increased finer timescale entropy (1-30 ms) and higher frequency band power (11-40 Hz) before training, but these differences disappeared following cognitive training. Our results suggest that in older adults with post-COVID syndrome, there is a pronounced shift from more global to local neural processing, potentially contributing to accelerated neural aging in this condition. However, cognitive training seems to offer a promising intervention method for modulating these changes in brain dynamics, especially among females.
Collapse
Affiliation(s)
- Boglárka Nagy
- Institute of Cognitive Neuroscience and Psychology, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary.
| | - Andrea B Protzner
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
- Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, Alberta, Canada
| | | | - Zsófia Anna Gaál
- Institute of Cognitive Neuroscience and Psychology, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
| |
Collapse
|
7
|
Wang S, Wang Y, Xu FH, Shen L, Zhao Y. Establishing group-level brain structural connectivity incorporating anatomical knowledge under latent space modeling. Med Image Anal 2024; 99:103309. [PMID: 39243600 DOI: 10.1016/j.media.2024.103309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 09/09/2024]
Abstract
Brain structural connectivity, capturing the white matter fiber tracts among brain regions inferred by diffusion MRI (dMRI), provides a unique characterization of brain anatomical organization. One fundamental question to address with structural connectivity is how to properly summarize and perform statistical inference for a group-level connectivity architecture, for instance, under different sex groups, or disease cohorts. Existing analyses commonly summarize group-level brain connectivity by a simple entry-wise sample mean or median across individual brain connectivity matrices. However, such a heuristic approach fully ignores the associations among structural connections and the topological properties of brain networks. In this project, we propose a latent space-based generative network model to estimate group-level brain connectivity. Within our modeling framework, we incorporate the anatomical information of brain regions as the attributes of nodes to enhance the plausibility of our estimation and improve biological interpretation. We name our method the attributes-informed brain connectivity (ABC) model, which compared with existing group-level connectivity estimations, (1) offers an interpretable latent space representation of the group-level connectivity, (2) incorporates the anatomical knowledge of nodes and tests its co-varying relationship with connectivity and (3) quantifies the uncertainty and evaluates the likelihood of the estimated group-level effects against chance. We devise a novel Bayesian MCMC algorithm to estimate the model. We evaluate the performance of our model through extensive simulations. By applying the ABC model to study brain structural connectivity stratified by sex among Alzheimer's Disease (AD) subjects and healthy controls incorporating the anatomical attributes (volume, thickness and area) on nodes, our method shows superior predictive power on out-of-sample structural connectivity and identifies meaningful sex-specific network neuromarkers for AD.
Collapse
Affiliation(s)
- Selena Wang
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, United States of America.
| | - Yiting Wang
- Department of Statistics, Virginia University, United States of America
| | - Frederick H Xu
- Department of Bioengineering, University of Pennsylvania, United States of America
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, United States of America
| | - Yize Zhao
- Department of Biostatistics, Yale Univeristy, United States of America
| |
Collapse
|
8
|
Kim SM, Sultana F, Korkmaz F, Rojekar S, Pallapati A, Ryu V, Lizneva D, Yuen T, Rosen CJ, Zaidi M. Neuroendocrinology of bone. Pituitary 2024:10.1007/s11102-024-01437-5. [PMID: 39096452 DOI: 10.1007/s11102-024-01437-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/26/2024] [Indexed: 08/05/2024]
Abstract
The past decade has witnessed significant advances in our understanding of skeletal homeostasis and the mechanisms that mediate the loss of bone in primary and secondary osteoporosis. Recent breakthroughs have primarily emerged from identifying disease-causing mutations and phenocopying human bone disease in rodents. Notably, using genetically-modified rodent models, disrupting the reciprocal relationship with tropic pituitary hormone and effector hormones, we have learned that pituitary hormones have independent roles in skeletal physiology, beyond their effects exerted through target endocrine glands. The rise of follicle-stimulating hormone (FSH) in the late perimenopause may account, at least in part, for the rapid bone loss when estrogen is normal, while low thyroid-stimulating hormone (TSH) levels may contribute to the bone loss in thyrotoxicosis. Admittedly speculative, suppressed levels of adrenocorticotropic hormone (ACTH) may directly exacerbate bone loss in the setting of glucocorticoid-induced osteoporosis. Furthermore, beyond their established roles in reproduction and lactation, oxytocin and prolactin may affect intergenerational calcium transfer and therefore fetal skeletal mineralization, whereas elevated vasopressin levels in chronic hyponatremic states may increase the risk of bone loss.. Here, we discuss the interaction of each pituitary hormone in relation to its role in bone physiology and pathophysiology.
Collapse
Affiliation(s)
- Se-Min Kim
- Mount Sinai Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Farhath Sultana
- Mount Sinai Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Funda Korkmaz
- Mount Sinai Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Satish Rojekar
- Mount Sinai Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Anusha Pallapati
- Mount Sinai Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Vitaly Ryu
- Mount Sinai Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Daria Lizneva
- Mount Sinai Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Tony Yuen
- Mount Sinai Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | | - Mone Zaidi
- Mount Sinai Center of Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
9
|
Lee BH, Cevizci M, Lieblich SE, Ibrahim M, Wen Y, Eid RS, Lamers Y, Duarte-Guterman P, Galea LAM. Exploring the parity paradox: Differential effects on neuroplasticity and inflammation by APOEe4 genotype at middle age. Brain Behav Immun 2024; 120:54-70. [PMID: 38772427 DOI: 10.1016/j.bbi.2024.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 04/20/2024] [Accepted: 05/18/2024] [Indexed: 05/23/2024] Open
Abstract
Female sex and Apolipoprotein E (APOE) ε4 genotype are top non-modifiable risk factors for Alzheimer's disease (AD). Although female-unique experiences like parity (pregnancy and motherhood) have positive effects on neuroplasticity at middle age, previous pregnancy may also contribute to AD risk. To explore these seemingly paradoxical long-term effects of parity, we investigated the impact of parity with APOEε4 genotype by examining behavioural and neural biomarkers of brain health in middle-aged female rats. Our findings show that primiparous (parous one time) hAPOEε4 rats display increased use of a non-spatial cognitive strategy and exhibit decreased number and recruitment of new-born neurons in the ventral dentate gyrus of the hippocampus in response to spatial working memory retrieval. Furthermore, primiparity and hAPOEε4 genotype synergistically modulate inflammatory markers in the ventral hippocampus. Collectively, these findings demonstrate that previous parity in hAPOEε4 rats confers an added risk to present with reduced activity and engagement of the hippocampus as well as elevated pro-inflammatory signaling, and underscore the importance of considering female-specific factors and genotype in health research.
Collapse
Affiliation(s)
- Bonnie H Lee
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Melike Cevizci
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Stephanie E Lieblich
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Muna Ibrahim
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Yanhua Wen
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Rand S Eid
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Yvonne Lamers
- Food Nutrition and Health Program, Faculty of Land and Food Systems, University of British Columbia, Vancouver, BC, Canada
| | - Paula Duarte-Guterman
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Liisa A M Galea
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, ON, Canada.
| |
Collapse
|
10
|
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
|
11
|
Ganesh A, Choudhury W, Coutellier L. Early spatial recognition memory deficits in 5XFAD female mice are associated with disruption of prefrontal parvalbumin neurons. Brain Res 2024; 1841:149122. [PMID: 39009061 DOI: 10.1016/j.brainres.2024.149122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/17/2024]
Abstract
Women have a two-fold increased risk of developing Alzheimer's disease (AD) than men, yet the underlying mechanisms of this sex-specific vulnerability remain unknown. Here, we aimed at determining in the 5XFAD mouse model whether deficits in prefrontal-dependent cognitive functions, which are impacted in the preclinical stages of AD, appear earlier in females, and whether these cognitive deficits are associated with alterations in the activity of prefrontal parvalbumin (PV)-neurons that regulate prefrontal circuits activity. We observed that 3.5-month-old 5XFAD females, but not males, display impairments in spatial short-term recognition memory, a function that relies on the integrity of the prefrontal cortex. Hippocampal-dependent cognitive functions were intact in both sexes. We then observed that 5XFAD females have more prefrontal PV neurons expressing the marker of chronic activity FosB; this was inversely correlated with prefrontal-dependent cognitive performances. Our findings show for the first time sex-specific, early deregulation of prefrontal PV neurons activity, which is associated with early appearance of prefrontal-dependent cognitive functions in 5XFAD females providing a potential novel mechanism to the increased risk to AD in females.
Collapse
Affiliation(s)
- Anish Ganesh
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Wajih Choudhury
- Department of Psychology, The Ohio State University, Columbus, OH, USA
| | - Laurence Coutellier
- Department of Psychology, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
12
|
Halloway S, Volgman AS, Schoeny ME, Arvanitakis Z, Barnes LL, Pressler SJ, Vispute S, Braun LT, Tafini S, Williams M, Wilbur J. Overcoming Pandemic-Related Challenges in Recruitment and Screening: Strategies and Representation of Older Women With Cardiovascular Disease for a Multidomain Lifestyle Trial to Prevent Cognitive Decline. J Cardiovasc Nurs 2024; 39:359-370. [PMID: 37167428 PMCID: PMC10638460 DOI: 10.1097/jcn.0000000000001000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
BACKGROUND Recruiting participants with cardiovascular disease into research during the COVID-19 pandemic was challenging, particularly those at risk of health disparities. OBJECTIVE During the pandemic, 12 cohorts of older women with cardiovascular disease were recruited from cardiology clinics into a lifestyle intervention trial to prevent cognitive decline. Objectives were to ( a ) describe the results of modified recruitment/screening strategies to overcome pandemic-related challenges and ( b ) evaluate differences in age, race, and ethnicity between patients recruited/randomized, recruited/not randomized (entered recruitment but not randomized because of being ineligible or not interested), and not recruited (clinic patients who met preliminary criteria but did not enter recruitment). METHODS This was a cross-sectional descriptive analysis. In-person study strategies proposed before the COVID-19 pandemic were modified before study onset (September 2020). Women 65 years or older with cardiovascular disease were recruited from cardiology clinics by clinicians, posted flyers, and letters mailed to patients randomly selected from electronic health record data extractions. Patients were classified as recruited/randomized, recruited/not randomized, and not recruited. RESULTS Of 5719 patients potentially eligible, 1689 patients entered recruitment via referral (49.1%), posted flyers (0.5%), or mailed letters (50.3%), and 253 patients were successfully recruited/randomized. Recruited/randomized participants were, on average, 72.4 years old (range, 65-90 years old), non-Hispanic White (54.2%), non-Hispanic Black (38.3%), Hispanic/Latinx (1.6%), and other/not reported (5.1%). The recruited/randomized group was significantly younger with fewer patients of Hispanic/Latinx ethnicity compared with those not recruited. CONCLUSIONS During the pandemic, all recruitment/screening goals were met using modified strategies. Differences in sociodemographic representation indicate a need for tailored strategies.
Collapse
|
13
|
Rehnberg J, Fors S, Ford KJ, Leist AK. Cognitive performance trends among European older adults: exploring variations across cohorts, gender, and educational levels (2007-2017). BMC Public Health 2024; 24:1646. [PMID: 38902637 PMCID: PMC11188163 DOI: 10.1186/s12889-024-19123-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/12/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND This study explores recent cohort trends in cognitive performance among older Europeans from 2007 to 2017, addressing three key questions: (1) Did cognitive performance improve universally and across the performance distribution during this period? (2) Did these improvements occur across educational levels and for both men and women? (3) Can established risk factors explain these performance gains? METHODS Using data from the Survey of Health, Ageing and Retirement in Europe (SHARE) across 12 European countries, we assessed immediate recall, delayed recall, and verbal fluency in individuals aged 60 to 94 in both 2007 and 2017 (n = 32 773). Differences between the two time points were estimated with linear mixed effects regression models and quantile regression. RESULTS Cognitive performance improved in all age groups, across educational levels, and for both men and women between 2007 and 2017. Notably, improvements were more pronounced at the upper end of the performance distribution for delayed recall and verbal fluency. Education explained approximately 20% of the observed improvements. Risk factors did not explain the observed improvements. CONCLUSIONS European cohorts of both younger-old and older adults continue to exhibit improvements in cognitive performance. Variation in the size of the cohort improvements across the performance distributions in delayed recall and in verbal fluency may contribute to growing inequalities in cognitive outcomes. Future research should further investigate the potential heterogeneity in cognitive performance gains. TRIAL REGISTRATION Not applicable.
Collapse
Affiliation(s)
- Johan Rehnberg
- Aging Research Center, Karolinska Institutet and Stockholm University, Tomtebodavägen 18A, Solna, Solna, SE-171 65, Sweden.
| | - Stefan Fors
- Aging Research Center, Karolinska Institutet and Stockholm University, Tomtebodavägen 18A, Solna, Solna, SE-171 65, Sweden
- Department of Public Health, Stockholm University, Albanovägen 12, Stockholm, Sweden
- Center for Epidemiology and Community Medicine, Region Stockholm, Stockholm, Solnavägen, 1E, Sweden
| | - Katherine J Ford
- Department of Psychology, Carleton University, Ottawa, K1S 5B6, Canada
| | - Anja K Leist
- Department of Social Sciences, University of Luxembourg, Esch-sur-Alzette, 4366, Luxembourg
| |
Collapse
|
14
|
Findley CA, McFadden S.A, Hill T, Peck MR, Quinn K, Hascup KN, Hascup ER. Sexual Dimorphism, Altered Hippocampal Glutamatergic Neurotransmission and Cognitive Impairment in APP Knock-In Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.05.570100. [PMID: 38106074 PMCID: PMC10723272 DOI: 10.1101/2023.12.05.570100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Background It is well established that glutamatergic neurotransmission plays an essential role in learning and memory. Previous studies indicate that glutamate dynamics shift with Alzheimer's disease (AD) progression, contributing to negative cognitive outcomes. Objective In this study, we characterized hippocampal glutamatergic signaling with age and disease progression in a knock-in mouse model of AD (APPNL-F/NL-F). Methods At 2-4 and 18+ months old, male and female APPNL/NL, APPNL-F/NL-F, and C57BL/6 mice underwent cognitive assessment using Morris water maze (MWM) and Novel Object Recognition (NOR). Then, basal and 70 mM KCl stimulus-evoked glutamate release was measured in the dentate gyrus (DG), CA3, and CA1 regions of the hippocampus using a glutamate-selective microelectrode in anesthetized mice. Results Glutamate recordings support elevated stimulus-evoked glutamate release in the DG and CA3 of young APPNL-F/NL-F male mice that declined with age compared to age-matched control mice. Young female APPNL-F/NL-F mice exhibited increased glutamate clearance in the CA1 that slowed with age compared to age-matched control mice. Male and female APPNL-F/NL-F mice exhibited decreased CA1 basal glutamate levels, while males also showed depletion in the CA3. Cognitive assessment demonstrated impaired spatial cognition in aged male and female APPNL-F/NL-F mice, but only aged females displayed recognition memory deficits compared to age-matched control mice. Conclusions: These findings confirm a sex-dependent hyper-to-hypoactivation glutamatergic paradigm in APPNL-F/NL-F mice. Further, data illustrate a sexually dimorphic biological aging process resulting in a more severe cognitive phenotype for female APPNL-F/NL-F mice than their male counterparts. Research outcomes mirror that of human AD pathology and provide further evidence of divergent AD pathogenesis between sexes.
Collapse
Affiliation(s)
- Caleigh A. Findley
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Depts of Neurology, Springfield, IL, USA
- Pharmacology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Samuel .A. McFadden
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Depts of Neurology, Springfield, IL, USA
| | - Tiarra. Hill
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Depts of Neurology, Springfield, IL, USA
| | - Mackenzie R. Peck
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Depts of Neurology, Springfield, IL, USA
| | - Kathleen Quinn
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Depts of Neurology, Springfield, IL, USA
| | - Kevin N. Hascup
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Depts of Neurology, Springfield, IL, USA
- Pharmacology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
- Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Erin R. Hascup
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Depts of Neurology, Springfield, IL, USA
- Pharmacology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| |
Collapse
|
15
|
Gong L, Liu D, Zhang B, Yu S, Xi C. Sex-Specific Entorhinal Cortex Functional Connectivity in Cognitively Normal Older Adults with Amyloid-β Pathology. Mol Neurobiol 2024:10.1007/s12035-024-04243-z. [PMID: 38867110 DOI: 10.1007/s12035-024-04243-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 05/06/2024] [Indexed: 06/14/2024]
Abstract
Sex and apolipoprotein E (APOE) genotype have been shown to influence the risk and progression of Alzheimer's disease (AD). However, the impact of these factors on the functional connectivity of the entorhinal cortex (ERC) in clinically unpaired older adults (CUOA) with amyloid-β (Aβ +) pathology remains unclear. A total of 1022 cognitively normal older adults with Aβ + (603 females and 586 APOE ε4 +) from the Anti-Amyloid Treatment in Asymptomatic Alzheimer's (A4) study were included in this study. The 2 × 2 (gender, 2 APOE genotypes) analysis of covariance was performed to compare the demographic information, cognitive performance, and volumetric MRI data among these groups. Voxel-wise comparisons of bilateral ERC functional connectivity (FC) were conducted, and partial correlation analyses were used to explore the associations between cognitive performance and ERC-FC strength. We found that the APOE genotype influenced ERC functional connectivity mainly in the sensorimotor network (SMN). Males exhibited higher ERC-FC in the salience network (SN), while females displayed higher ERC-FC in the default mode network (DMN), executive control network (ECN), and reward network. The interplay of sex and APOE genotype on ERC-FC was observed in the SMN and cerebellar lobe. The ERC-FC was associated with executive function and memory performance in individuals with CUOA-Aβ + . Our findings provide evidence of sex-specific ERC functional connectivity compensation mechanism in cognitively normal older adults with Aβ + pathology. This study may contribute to a better understanding of the mechanisms underlying the early stages of AD and may help develop personalized interventions in preclinical AD.
Collapse
Affiliation(s)
- Liang Gong
- Department of Neurology, Chengdu Second People's Hospital, Chengdu, 610017, Sichuan, China
| | - Duan Liu
- Department of Neurology, Chengdu Second People's Hospital, Chengdu, 610017, Sichuan, China
| | - Bei Zhang
- Department of Neurology, Chengdu Second People's Hospital, Chengdu, 610017, Sichuan, China
| | - Siyi Yu
- Department of Acupuncture & Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China.
| | - Chunhua Xi
- Department of Neurology, The Third Affiliated Hospital of Anhui Medical University, Huaihe Road 390, Heifei, 230061, Anhui, China.
| |
Collapse
|
16
|
Rikos N, Linardakis M, Smpokos E, Spiridaki E, Symvoulakis EK, Tsiligianni I, Philalithis A. Assessment of Cognitive Function in European Adults Aged 50+in Relation to Their Handgrip Strength and Physical Inactivity: The SHARE Study During 2019-2020. J Res Health Sci 2024; 24:e00611. [PMID: 39072547 PMCID: PMC11264452 DOI: 10.34172/jrhs.2024.146] [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: 08/06/2023] [Revised: 08/27/2023] [Accepted: 04/23/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND Cognitive function is crucial during aging. This study assessed the cognitive function of European adults aged 50 and over in relation to handgrip strength and physical inactivity. Study Design: This was a cross-sectional survey. METHODS Data were collected from 41,395 adults from 27 European countries participating in the Survey of Health, Ageing, and Retirement in Europe (SHARE) during 2019-2020. Cognitive function was assessed based on five tests, and cognitive impairment was defined using 3+tests. Handgrip strength and physical inactivity were also correlated through the analysis of covariance using a complex study design. RESULTS The majority of participants were female (56.6%), with a mean age of 70.9 years, and 22.6% presented multimorbidity. Furthermore, 51.1% had a normal cognitive function, while 13.3% had cognitive impairment (The estimated population was 21,944,722). Moreover, cognitive impairment was more prevalent in females than in males (14.4% vs. 12.0%, P<0.001) in patients with no years of education (P<0.001) and origin from southern European countries (P<0.001). Additionally, participants with cognitive impairment had lower mean handgrip strength compared to those with cognitive impairment in 1-2 criteria or with normal cognitive function (29.3 vs. 33.4 and 35.1 kg, respectively, P<0.001). Physically inactive participants had higher odds ratio (OR) of cognitive impairment than those engaging in moderate/vigorous physical activity, both in 1-2 tests (OR:1.73, 95% confidence interval (CI): 1.32-2.26) and in 3+tests (OR: 3.36, 95% CI: 2.57-4.40). CONCLUSION Cognitive impairment presented low prevalence and was associated with low levels of handgrip strength and physical inactivity. These specific factors may play a special role in early detection, diagnosis, and treatment or may slow down the progression of cognitive impairment.
Collapse
Affiliation(s)
- Nikos Rikos
- Department of Nursing, School of Health Sciences, Hellenic Mediterranean University, Heraklion, Greece
| | - Manolis Linardakis
- Department of Social Medicine, Faculty of Medicine, University of Crete, Greece
| | - Emmanouil Smpokos
- Department of Social Medicine, Faculty of Medicine, University of Crete, Greece
| | - Eleni Spiridaki
- Department of Social Medicine, Faculty of Medicine, University of Crete, Greece
| | | | - Ioanna Tsiligianni
- Department of Social Medicine, Faculty of Medicine, University of Crete, Greece
| | - Anastas Philalithis
- Department of Social Medicine, Faculty of Medicine, University of Crete, Greece
| |
Collapse
|
17
|
Onisiforou A, Christodoulou CC, Zamba-Papanicolaou E, Zanos P, Georgiou P. Transcriptomic analysis reveals sex-specific patterns in the hippocampus in Alzheimer's disease. Front Endocrinol (Lausanne) 2024; 15:1345498. [PMID: 38689734 PMCID: PMC11058985 DOI: 10.3389/fendo.2024.1345498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 04/02/2024] [Indexed: 05/02/2024] Open
Abstract
Background The hippocampus, vital for memory and learning, is among the first brain regions affected in Alzheimer's Disease (AD) and exhibits adult neurogenesis. Women face twice the risk of developing AD compare to men, making it crucial to understand sex differences in hippocampal function for comprehending AD susceptibility. Methods We conducted a comprehensive analysis of bulk mRNA postmortem samples from the whole hippocampus (GSE48350, GSE5281) and its CA1 and CA3 subfields (GSE29378). Our aim was to perform a comparative molecular signatures analysis, investigating sex-specific differences and similarities in the hippocampus and its subfields in AD. This involved comparing the gene expression profiles among: (a) male controls (M-controls) vs. female controls (F-controls), (b) females with AD (F-AD) vs. F-controls, (c) males with AD (M-AD) vs. M-controls, and (d) M-AD vs. F-AD. Furthermore, we identified AD susceptibility genes interacting with key targets of menopause hormone replacement drugs, specifically the ESR1 and ESR2 genes, along with GPER1. Results The hippocampal analysis revealed contrasting patterns between M-AD vs. M-controls and F-AD vs. F-controls, as well as M-controls vs. F-controls. Notably, BACE1, a key enzyme linked to amyloid-beta production in AD pathology, was found to be upregulated in M-controls compared to F-controls in both CA1 and CA3 hippocampal subfields. In M-AD vs. M-controls, the GABAergic synapse was downregulated, and the Estrogen signaling pathway was upregulated in both subfields, unlike in F-AD vs. F-controls. Analysis of the whole hippocampus also revealed upregulation of the GABAergic synapse in F-AD vs. F-controls. While direct comparison of M-AD vs. F-AD, revealed a small upregulation of the ESR1 gene in the CA1 subfield of males. Conversely, F-AD vs. F-controls exhibited downregulation of the Dopaminergic synapse in both subfields, while the Calcium signaling pathway showed mixed regulation, being upregulated in CA1 but downregulated in CA3, unlike in M-AD vs. M-controls. The upregulated Estrogen signaling pathway in M-AD, suggests a compensatory response to neurodegenerative specifically in males with AD. Our results also identified potential susceptibility genes interacting with ESR1 and ESR2, including MAPK1, IGF1, AKT1, TP53 and CD44. Conclusion These findings underscore the importance of sex-specific disease mechanisms in AD pathogenesis. Region-specific analysis offers a more detailed examination of localized changes in the hippocampus, enabling to capture sex-specific molecular patterns in AD susceptibility and progression.
Collapse
Affiliation(s)
- Anna Onisiforou
- Translational Neuropharmacology Laboratory, Department of Psychology, University of Cyprus, Nicosia, Cyprus
| | | | | | - Panos Zanos
- Translational Neuropharmacology Laboratory, Department of Psychology, University of Cyprus, Nicosia, Cyprus
| | - Polymnia Georgiou
- Laboratory of Epigenetics and Gene Regulation, Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
- Psychoneuroendocrinology Laboratory, Department of Psychology, University of Wisconsin Milwaukee, Milwaukee, WI, United States
| |
Collapse
|
18
|
Barth C, Galea LA, Jacobs EG, Lee BH, Westlye LT, de Lange AMG. Menopausal hormone therapy and the female brain: leveraging neuroimaging and prescription registry data from the UK Biobank cohort. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.08.24305450. [PMID: 38645009 PMCID: PMC11030497 DOI: 10.1101/2024.04.08.24305450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Background and Objectives Menopausal hormone therapy (MHT) is generally thought to be neuroprotective, yet results have been inconsistent. Here, we present a comprehensive study of MHT use and brain characteristics in middle- to older aged females from the UK Biobank, assessing detailed MHT data, APOE ε4 genotype, and tissue-specific gray (GM) and white matter (WM) brain age gap (BAG), as well as hippocampal and white matter hyperintensity (WMH) volumes. Methods A total of 19,846 females with magnetic resonance imaging data were included (current-users = 1,153, 60.1 ± 6.8 years; past-users = 6,681, 67.5 ± 6.2 years; never-users = 12,012, mean age 61.6 ± 7.1 years). For a sub-sample (n = 538), MHT prescription data was extracted from primary care records. Brain measures were derived from T1-, T2- and diffusion-weighted images. We fitted regression models to test for associations between the brain measures and MHT variables including user status, age at initiation, dosage and duration, formulation, route of administration, and type (i.e., bioidentical vs synthetic), as well as active ingredient (e.g., estradiol hemihydrate). We further tested for differences in brain measures among MHT users with and without a history of hysterectomy ± bilateral oophorectomy and examined associations by APOE ε4 status. Results We found significantly higher GM and WM BAG (i.e., older brain age relative to chronological age) as well as smaller left and right hippocampus volumes in current MHT users, not past users, compared to never-users. Effects were modest, with the largest effect size indicating a group difference of 0.77 years (~9 months) for GM BAG. Among MHT users, we found no significant associations between age at MHT initiation and brain measures. Longer duration of use and older age at last use post menopause was associated with higher GM and WM BAG, larger WMH volume, and smaller left and right hippocampal volumes. MHT users with a history of hysterectomy ± bilateral oophorectomy showed lower GM BAG relative to MHT users without such history. Although we found smaller hippocampus volumes in carriers of two APOE ε4 alleles compared to non-carriers, we found no interactions with MHT variables. In the sub-sample with prescription data, we found no significant associations between detailed MHT variables and brain measures after adjusting for multiple comparisons. Discussion Our results indicate that population-level associations between MHT use, and female brain health might vary depending on duration of use and past surgical history. Future research is crucial to establish causality, dissect interactions between menopause-related neurological changes and MHT use, and determine individual-level implications to advance precision medicine in female health care.
Collapse
Affiliation(s)
- Claudia Barth
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Liisa A.M. Galea
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Emily G. Jacobs
- Psychological and Brain Sciences, University of California Santa Barbara, CA, USA
| | - Bonnie H. Lee
- Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Lars T. Westlye
- Department of Psychology, University of Oslo, Oslo, Norway
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Ann-Marie G. de Lange
- Department of Psychology, University of Oslo, Oslo, Norway
- Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
- Department of Psychiatry, University of Oxford, Oxford, UK
| |
Collapse
|
19
|
Nguyen CQN, Ma L, Low YLC, Tan ECK, Fowler C, Masters CL, Jin L, Pan Y. Exploring the link between comorbidities and Alzheimer's dementia in the Australian Imaging, Biomarker & Lifestyle (AIBL) study. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12593. [PMID: 38770381 PMCID: PMC11103763 DOI: 10.1002/dad2.12593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/26/2024] [Accepted: 03/30/2024] [Indexed: 05/22/2024]
Abstract
INTRODUCTION Mounting evidence suggests that certain comorbidities may influence the clinical evolution of Alzheimer's dementia (AD). METHODS We conducted logistic regression analyses on the medical history and cognitive health diagnoses of participants in the Australian Imaging, Biomarker & Lifestyle study (n = 2443) to investigate cross-sectional associations between various comorbidities and mild cognitive impairment (MCI)/AD. RESULTS A mixture of associations were observed. Higher comorbidity of anxiety and other neurological disorders was associated with higher odds of AD, while arthritis, cancer, gastric complaints, high cholesterol, joint replacement, visual defect, kidney and liver disease were associated with lower odds of AD. DISCUSSION This study underscores the links between specific comorbidities and MCI/AD. Further research is needed to elucidate the longitudinal comorbidity-MCI/AD associations and underlying mechanisms of these associations. Highlights Comorbidities that significantly increased AD odds included anxiety and other neurological disorders.Arthritis, cancer, gastric complaints, high cholesterol, joint replacement, visual defect, kidney and liver disease were associated with lower odds of AD.Alcohol consumption had the most significant confounding effect in the study.Visual-AD association was modified by age, sex, and APOE ε4 allele status.Anxiety-AD and depression-AD associations were modified by sex.
Collapse
Affiliation(s)
| | - Liwei Ma
- The Florey InstituteThe University of MelbourneParkvilleVictoriaAustralia
| | - Yi Ling Clare Low
- The Florey InstituteThe University of MelbourneParkvilleVictoriaAustralia
| | - Edwin C. K. Tan
- School of PharmacyFaculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
| | - Christopher Fowler
- The Florey InstituteThe University of MelbourneParkvilleVictoriaAustralia
| | - Colin L. Masters
- The Florey InstituteThe University of MelbourneParkvilleVictoriaAustralia
| | - Liang Jin
- The Florey InstituteThe University of MelbourneParkvilleVictoriaAustralia
| | - Yijun Pan
- The Florey InstituteThe University of MelbourneParkvilleVictoriaAustralia
- Drug Delivery, Disposition and DynamicsMonash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVictoriaAustralia
- Department of Organ AnatomyGraduate School of MedicineTohoku UniversitySendaiMiyagiJapan
| | | |
Collapse
|
20
|
El Haj M, Allain P, Boutoleau-Bretonnière C, Chapelet G, Kapogiannis D, Ndobo A. Does Sex Matter? High Semantic Autobiographical Retrieval in Women and Men With Alzheimer's Disease. Psychol Rep 2024; 127:649-667. [PMID: 36165092 PMCID: PMC10040469 DOI: 10.1177/00332941221130223] [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] [Indexed: 10/14/2022]
Abstract
The decline of autobiographical memory in Alzheimer's disease (AD) is mainly characterized by overgenerality. While there is a large body of research on autobiographical overgenerality in AD, this research has mainly assessed retrieval with a dichotomy between specific vs. general retrieval. To go beyond this dichotomy, we assessed several degrees of autobiographical specificity in patients with AD, namely, we assessed specific vs. categoric vs. extended vs. semantic retrieval. We also assessed sex differences regarding these degrees of autobiographical specificity. We invited patients with mild AD and control participants to complete sentences (e. g., "When I think back to/of…") with autobiographical memories. Memories were categorized into specific, categoric, extended, or semantic memories. Results demonstrated more semantic than specific, categoric or extended memories in men and women with AD. In control participants, analysis demonstrated more specific than categoric, extended, and semantic memories in men and women. Also, no significant differences were observed between women and men with AD, or between control women and men, regarding specific, categoric, extended, and semantic memoires. This study offers not only a nuanced analysis of autobiographical specificity in patients with mild AD, but also an original analysis regarding this specificity by sex.
Collapse
Affiliation(s)
- Mohamad El Haj
- Nantes Université, Univ Angers, Laboratoire de psychologie des Pays de la Loire, Nantes, France; CHU Nantes, Clinical Gerontology Department, Bd Jacques Monod, Nantes, France; Institut Universitaire de France, Paris, France
| | - Philippe Allain
- Laboratoire de Psychologie des Pays de la Loire, LPPL EA 4638, SFR Confluences, UNIV Angers, Nantes Université, Maison de la recherche Germaine Tillion, Angers, France; Département de Neurologie, CHU Angers, Angers, France
| | - Claire Boutoleau-Bretonnière
- CHU Nantes, Inserm CIC04, Nantes, France; CHU Nantes, Département de Neurologie, Centre Mémoire de Ressources et Recherche, Nantes, France
| | - Guillaume Chapelet
- Université de Nantes, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, Nantes, France; CHU Nantes, Clinical Gerontology Department, Bd Jacques Monod, Nantes, France
| | - Dimitrios Kapogiannis
- Laboratory of Clinical Investigation, National Institute on Aging, Baltimore, MD, USA
| | - André Ndobo
- Nantes Université, Univ Angers, Laboratoire de psychologie des Pays de la Loire, Nantes, France
| |
Collapse
|
21
|
Jockwitz C, Krämer C, Dellani P, Caspers S. Differential predictability of cognitive profiles from brain structure in older males and females. GeroScience 2024; 46:1713-1730. [PMID: 37730943 PMCID: PMC10828131 DOI: 10.1007/s11357-023-00934-y] [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: 07/27/2023] [Accepted: 09/04/2023] [Indexed: 09/22/2023] Open
Abstract
Structural brain imaging parameters may successfully predict cognitive performance in neurodegenerative diseases but mostly fail to predict cognitive abilities in healthy older adults. One important aspect contributing to this might be sex differences. Behaviorally, older males and females have been found to differ in terms of cognitive profiles, which cannot be captured by examining them as one homogenous group. In the current study, we examined whether the prediction of cognitive performance from brain structure, i.e. region-wise grey matter volume (GMV), would benefit from the investigation of sex-specific cognitive profiles in a large sample of older adults (1000BRAINS; N = 634; age range 55-85 years). Prediction performance was assessed using a machine learning (ML) approach. Targets represented a) a whole-sample cognitive component solution extracted from males and females, and b) sex-specific cognitive components. Results revealed a generally low predictability of cognitive profiles from region-wise GMV. In males, low predictability was observed across both, the whole sample as well as sex-specific cognitive components. In females, however, predictability differences across sex-specific cognitive components were observed, i.e. visual working memory (WM) and executive functions showed higher predictability than fluency and verbal WM. Hence, results accentuated that addressing sex-specific cognitive profiles allowed a more fine-grained investigation of predictability differences, which may not be observable in the prediction of the whole-sample solution. The current findings not only emphasize the need to further investigate the predictive power of each cognitive component, but they also emphasize the importance of sex-specific analyses in older adults.
Collapse
Affiliation(s)
- Christiane Jockwitz
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany.
- Institute for Anatomy I, Medical Faculty & University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| | - Camilla Krämer
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
- Institute for Anatomy I, Medical Faculty & University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Paulo Dellani
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
- Institute for Anatomy I, Medical Faculty & University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Svenja Caspers
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, Jülich, Germany
- Institute for Anatomy I, Medical Faculty & University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
22
|
Lv Y, Su H, Li R, Yang Z, Chen Q, Zhang D, Liang S, Hu C, Ni X. A cross-sectional study of the major risk factor at different levels of cognitive performance within Chinese-origin middle-aged and elderly individuals. J Affect Disord 2024; 349:377-383. [PMID: 38199420 DOI: 10.1016/j.jad.2024.01.069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 12/20/2023] [Accepted: 01/04/2024] [Indexed: 01/12/2024]
Abstract
OBJECTIVE Senior citizens suffering from cognitive impairment (CI) are on the East Asia rise. Multiple variables could lead to inter-/intra-individual cognition effectiveness variations, though previous research efforts did not consider weighting issues. METHODS This study scrutinized 5639 participants meeting required inclusion criteria by the CHARLS. Cognitive capacity was evaluated through Mini-Mental State Examination (MMSE). Considering that MMSE scorings were not following normal distribution, a non-parametric test and multiple linear regression were performed to screen candidate variables linked to cognitive capacity. Such applicability of candidate factors in the cumulative effect and the weighting of the impact on cognitive performance were evaluated by random forest (RF) algorithm. RESULTS Age, gender, education, marital status, residence, the type of residence, exercise, socialization level and drinking were correlated to MMSE scorings (p < 0.05). Among them, age, education, gender and sociality were correlated to individual MMSE items (p < 0.05). Regardless of MMSE scores and several MMSE items, age is always a prime factor. However, in the attention and computation item, education is better than age and ranks first. CONCLUSIONS This preliminary study prompted age, education, gender, and sociality with varying weightings to be linked to cognitive capacity within a Chinese cohort by differing cognitive aspects. At different levels of cognitive performance, the main risk factors are basically similar, but there are still some differences.
Collapse
Affiliation(s)
- Yuan Lv
- Jiangbin Hospital of Guangxi Zhuang Autonomous Region, 530021, PR China
| | - Huabin Su
- Jiangbin Hospital of Guangxi Zhuang Autonomous Region, 530021, PR China
| | - Rongqiao Li
- Jiangbin Hospital of Guangxi Zhuang Autonomous Region, 530021, PR China
| | - Ze Yang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, 100730, PR China
| | - Qing Chen
- Jiangbin Hospital of Guangxi Zhuang Autonomous Region, 530021, PR China
| | - Di Zhang
- Jiangbin Hospital of Guangxi Zhuang Autonomous Region, 530021, PR China
| | - Shuolin Liang
- Jiangbin Hospital of Guangxi Zhuang Autonomous Region, 530021, PR China
| | - Caiyou Hu
- Jiangbin Hospital of Guangxi Zhuang Autonomous Region, 530021, PR China
| | - Xiaolin Ni
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China.
| |
Collapse
|
23
|
Khan N, Uribe Isaza J, Rouhi N, Jamani NF, Jabeen S, Gill AK, Tsutsui M, Visser F, Sargin D. Behavioral and Neurophysiological Implications of Pathological Human Tau Expression in Serotonin Neurons. ACS Chem Neurosci 2024; 15:932-943. [PMID: 38377680 PMCID: PMC10921395 DOI: 10.1021/acschemneuro.3c00626] [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: 09/28/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/22/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive degenerative disorder that results in a severe loss of brain cells and irreversible cognitive decline. Memory problems are the most recognized symptoms of AD. However, approximately 90% of patients diagnosed with AD suffer from behavioral symptoms, including mood changes and social impairment years before cognitive dysfunction. Recent evidence indicates that the dorsal raphe nucleus (DRN) is among the initial regions that show tau pathology, which is a hallmark feature of AD. The DRN harbors serotonin (5-HT) neurons, which are critically involved in mood, social, and cognitive regulation. Serotonergic impairment early in the disease process may contribute to behavioral symptoms in AD. However, the mechanisms underlying vulnerability and contribution of the 5-HT system to AD progression remain unknown. Here, we performed behavioral and electrophysiological characterizations in mice expressing a phosphorylation-prone form of human tau (hTauP301L) in 5-HT neurons. We found that pathological tau expression in 5-HT neurons induces anxiety-like behavior and alterations in stress-coping strategies in female and male mice. Female mice also exhibited social disinhibition and mild cognitive impairment in response to 5-HT neuron-specific hTauP301L expression. Behavioral alterations were accompanied by disrupted 5-HT neuron physiology in female and male hTauP301L expressing mice with exacerbated excitability disruption in females only. These data provide mechanistic insights into the brain systems and symptoms impaired early in AD progression, which is critical for disease intervention.
Collapse
Affiliation(s)
- Nazmus
S. Khan
- Department
of Psychology, Department of Physiology and Pharmacology, Cumming School of
Medicine, Hotchkiss Brain Institute, Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Juan Uribe Isaza
- Department
of Psychology, Department of Physiology and Pharmacology, Cumming School of
Medicine, Hotchkiss Brain Institute, Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Nahid Rouhi
- Department
of Psychology, Department of Physiology and Pharmacology, Cumming School of
Medicine, Hotchkiss Brain Institute, Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Naila F. Jamani
- Department
of Psychology, Department of Physiology and Pharmacology, Cumming School of
Medicine, Hotchkiss Brain Institute, Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Shaista Jabeen
- Department
of Psychology, Department of Physiology and Pharmacology, Cumming School of
Medicine, Hotchkiss Brain Institute, Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Amisha K. Gill
- Department
of Psychology, Department of Physiology and Pharmacology, Cumming School of
Medicine, Hotchkiss Brain Institute, Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Mio Tsutsui
- Department
of Psychology, Department of Physiology and Pharmacology, Cumming School of
Medicine, Hotchkiss Brain Institute, Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Frank Visser
- Department
of Psychology, Department of Physiology and Pharmacology, Cumming School of
Medicine, Hotchkiss Brain Institute, Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Derya Sargin
- Department
of Psychology, Department of Physiology and Pharmacology, Cumming School of
Medicine, Hotchkiss Brain Institute, Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| |
Collapse
|
24
|
Liu J, Zhang Y, Jia F, Zhang H, Luo L, Liao Y, Ouyang M, Yi X, Zhu R, Bai W, Ning G, Li X, Qu H. Sex differences in fetal brain functional network topology. Cereb Cortex 2024; 34:bhae111. [PMID: 38517172 DOI: 10.1093/cercor/bhae111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 03/23/2024] Open
Abstract
The fetal period is a critical stage in brain development, and understanding the characteristics of the fetal brain is crucial. Although some studies have explored aspects of fetal brain functional networks, few have specifically focused on sex differences in brain network characteristics. We adopted the graph theory method to calculate brain network functional connectivity and topology properties (including global and nodal properties), and further compared the differences in these parameters between male and female fetuses. We found that male fetuses showed an increased clustering coefficient and local efficiency than female fetuses, but no significant group differences concerning other graph parameters and the functional connectivity matrix. Our study suggests the existence of sex-related distinctions in the topological properties of the brain network at the fetal stage of development and demonstrates an increase in brain network separation in male fetuses compared with female fetuses.
Collapse
Affiliation(s)
- Jing Liu
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, P.R. China
- Ministry of Education, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Chengdu 610041, Sichuan, P.R. China
| | - Yujin Zhang
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, P.R. China
- Ministry of Education, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Chengdu 610041, Sichuan, P.R. China
| | - Fenglin Jia
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, P.R. China
- Ministry of Education, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Chengdu 610041, Sichuan, P.R. China
| | - Hongding Zhang
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, P.R. China
- Ministry of Education, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Chengdu 610041, Sichuan, P.R. China
| | - Lekai Luo
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, P.R. China
- Ministry of Education, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Chengdu 610041, Sichuan, P.R. China
| | - Yi Liao
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, P.R. China
- Ministry of Education, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Chengdu 610041, Sichuan, P.R. China
| | - Minglei Ouyang
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, P.R. China
- Ministry of Education, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Chengdu 610041, Sichuan, P.R. China
| | - Xiaoxue Yi
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, P.R. China
- Ministry of Education, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Chengdu 610041, Sichuan, P.R. China
| | - Ruixi Zhu
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, P.R. China
- Ministry of Education, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Chengdu 610041, Sichuan, P.R. China
| | - Wanjing Bai
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, P.R. China
- Ministry of Education, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Chengdu 610041, Sichuan, P.R. China
| | - Gang Ning
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, P.R. China
- Ministry of Education, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Chengdu 610041, Sichuan, P.R. China
| | - Xuesheng Li
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, P.R. China
- Ministry of Education, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Chengdu 610041, Sichuan, P.R. China
| | - Haibo Qu
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, P.R. China
- Ministry of Education, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Chengdu 610041, Sichuan, P.R. China
| |
Collapse
|
25
|
Frolinger T, Korkmaz F, Sims S, Sen F, Sultana F, Laurencin V, Cullen L, Pallapati AR, Liu A, Rojekar S, Pevnev G, Cheliadinova U, Vasilyeva D, Burganova G, Macdonald A, Saxena M, Goosens K, Rosen C, Barak O, Lizneva D, Gumerova A, Ye K, Ryu V, Yuen T, Zaidi M. Gene-Dose-Dependent Reduction Fshr Expression Improves Spatial Memory Deficits in Alzheimer's Mice. RESEARCH SQUARE 2024:rs.3.rs-3964789. [PMID: 38463956 PMCID: PMC10925392 DOI: 10.21203/rs.3.rs-3964789/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Alzheimer's disease (AD) is a major progressive neurodegenerative disorder of the aging population. High post-menopausal levels of the pituitary gonadotropin follicle-stimulating hormone (FSH) are strongly associated with the onset of AD, and we have shown recently that FSH directly activates the hippocampal Fshr to drive AD-like pathology and memory loss in mice. To establish a role for FSH in memory loss, we used female 3xTg;Fshr+/+, 3xTg;Fshr+/- and 3xTg;Fshr-/- mice that were either left unoperated or underwent sham surgery or ovariectomy at 8 weeks of age. Unoperated and sham-operated 3xTg;Fshr-/- mice were implanted with 17β-estradiol pellets to normalize estradiol levels. Morris Water Maze and Novel Object Recognition behavioral tests were performed to study deficits in spatial and recognition memory, respectively, and to examine the effects of Fshr depletion. 3xTg;Fshr+/+ mice displayed impaired spatial memory at 5 months of age; both the acquisition and retrieval of the memory were ameliorated in 3xTg;Fshr-/- mice and, to a lesser extent, in 3xTg;Fshr+/- mice- -thus documenting a clear gene-dose-dependent prevention of hippocampal-dependent spatial memory impairment. At 5 and 10 months, sham-operated 3xTg;Fshr-/- mice showed better memory performance during the acquasition and/or retrieval phases, suggesting that Fshr deletion prevented the progression of spatial memory deficits with age. However, this prevention was not seen when mice were ovariectomized, except in the 10-month-old 3xTg;Fshr-/- mice. In the Novel Object Recognition test performed at 10 months, all groups of mice, except ovariectomized 3xTg;Fshr-/- mice showed a loss of recognition memory. Consistent with the neurobehavioral data, there was a gene-dose-dependent reduction mainly in the amyloid β40 isoform in whole brain extracts. Finally, serum FSH levels < 8 ng/mL in 16-month-old APP/PS1 mice were associated with better retrieval of spatial memory. Collectively, the data provide compelling genetic evidence for a protective effect of inhibiting FSH signaling on the progression of spatial and recognition memory deficits in mice, and lay a firm foundation for the use of an FSH-blocking agent for the early prevention of cognitive decline in postmenopausal women.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Avi Liu
- Icahn School of Medicine at Mount Sinai
| | | | | | | | | | | | | | | | | | | | | | | | | | - Keqiang Ye
- Shenzhen Institute of Advanced Technology
| | | | | | | |
Collapse
|
26
|
Ferretti MT, Ding H, Au R, Liu C, Devine S, Auerbach S, Mez J, Gurnani A, Liu Y, Santuccione A, Ang TFA. Maximizing utility of neuropsychological measures in sex-specific predictive models of incident Alzheimer's disease in the Framingham Heart Study. Alzheimers Dement 2024; 20:1112-1122. [PMID: 37882354 PMCID: PMC10917035 DOI: 10.1002/alz.13500] [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: 02/21/2023] [Revised: 08/23/2023] [Accepted: 09/17/2023] [Indexed: 10/27/2023]
Abstract
INTRODUCTION Sex differences in neuropsychological (NP) test performance might have important implications for the diagnosis of Alzheimer's disease (AD). This study investigates sex differences in neuropsychological performance among individuals without dementia at baseline. METHODS Neuropsychological assessment data, both standard test scores and process coded responses, from Framingham Heart Study participants were analyzed for sex differences using regression model and Cox proportional hazards model. Optimal NP profiles were identified by machine learning methods for men and women. RESULTS Sex differences were observed in both summary scores and composite process scores of NP tests in terms of adjusted means and their associations with AD incidence. The optimal NP profiles for men and women have 10 and 8 measures, respectively, and achieve 0.76 mean area under the curve for AD prediction. DISCUSSION These results suggest that NP tests can be leveraged for developing more sensitive, sex-specific indices for the diagnosis of AD.
Collapse
Affiliation(s)
- Maria Teresa Ferretti
- Institute for Regenerative Medicine (IREM)University of ZurichZurichSwitzerland
- Women's Brain ProjectGuntershausenSwitzerland
| | - Huitong Ding
- Department of Anatomy and NeurobiologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- The Framingham Heart StudyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Rhoda Au
- Department of Anatomy and NeurobiologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- The Framingham Heart StudyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Department of EpidemiologyBoston University School of Public HealthBostonMassachusettsUSA
- Slone Epidemiology CenterBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Department of NeurologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Chunyu Liu
- The Framingham Heart StudyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
| | - Sherral Devine
- Department of Anatomy and NeurobiologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- The Framingham Heart StudyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Sanford Auerbach
- Department of NeurologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Jesse Mez
- Department of NeurologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Ashita Gurnani
- Department of NeurologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | - Yulin Liu
- Department of Anatomy and NeurobiologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- The Framingham Heart StudyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| | | | - Ting Fang Alvin Ang
- Department of Anatomy and NeurobiologyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- The Framingham Heart StudyBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
- Slone Epidemiology CenterBoston University Chobanian & Avedisian School of MedicineBostonMassachusettsUSA
| |
Collapse
|
27
|
Biechele G, Rauchmann BS, Janowitz D, Buerger K, Franzmeier N, Weidinger E, Guersel S, Schuster S, Finze A, Harris S, Lindner S, Albert NL, Wetzel C, Rupprecht R, Rominger A, Palleis C, Katzdobler S, Burow L, Kurz C, Zaganjori M, Trappmann LK, Goldhardt O, Grimmer T, Haeckert J, Keeser D, Stoecklein S, Morenas-Rodriguez E, Bartenstein P, Levin J, Höglinger GU, Simons M, Perneczky R, Brendel M. Associations between sex, body mass index and the individual microglial response in Alzheimer's disease. J Neuroinflammation 2024; 21:30. [PMID: 38263017 PMCID: PMC10804830 DOI: 10.1186/s12974-024-03020-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/11/2024] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND AND OBJECTIVES 18-kDa translocator protein position-emission-tomography (TSPO-PET) imaging emerged for in vivo assessment of neuroinflammation in Alzheimer's disease (AD) research. Sex and obesity effects on TSPO-PET binding have been reported for cognitively normal humans (CN), but such effects have not yet been systematically evaluated in patients with AD. Thus, we aimed to investigate the impact of sex and obesity on the relationship between β-amyloid-accumulation and microglial activation in AD. METHODS 49 patients with AD (29 females, all Aβ-positive) and 15 Aβ-negative CN (8 female) underwent TSPO-PET ([18F]GE-180) and β-amyloid-PET ([18F]flutemetamol) imaging. In 24 patients with AD (14 females), tau-PET ([18F]PI-2620) was additionally available. The brain was parcellated into 218 cortical regions and standardized-uptake-value-ratios (SUVr, cerebellar reference) were calculated. Per region and tracer, the regional increase of PET SUVr (z-score) was calculated for AD against CN. The regression derived linear effect of regional Aβ-PET on TSPO-PET was used to determine the Aβ-plaque-dependent microglial response (slope) and the Aβ-plaque-independent microglial response (intercept) at the individual patient level. All read-outs were compared between sexes and tested for a moderation effect of sex on associations with body mass index (BMI). RESULTS In AD, females showed higher mean cortical TSPO-PET z-scores (0.91 ± 0.49; males 0.30 ± 0.75; p = 0.002), while Aβ-PET z-scores were similar. The Aβ-plaque-independent microglial response was stronger in females with AD (+ 0.37 ± 0.38; males with AD - 0.33 ± 0.87; p = 0.006), pronounced at the prodromal stage. On the contrary, the Aβ-plaque-dependent microglial response was not different between sexes. The Aβ-plaque-independent microglial response was significantly associated with tau-PET in females (Braak-II regions: r = 0.757, p = 0.003), but not in males. BMI and the Aβ-plaque-independent microglial response were significantly associated in females (r = 0.44, p = 0.018) but not in males (BMI*sex interaction: F(3,52) = 3.077, p = 0.005). CONCLUSION While microglia response to fibrillar Aβ is similar between sexes, women with AD show a stronger Aβ-plaque-independent microglia response. This sex difference in Aβ-independent microglial activation may be associated with tau accumulation. BMI is positively associated with the Aβ-plaque-independent microglia response in females with AD but not in males, indicating that sex and obesity need to be considered when studying neuroinflammation in AD.
Collapse
Affiliation(s)
- Gloria Biechele
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Boris-Stephan Rauchmann
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
- Institute of Neuroradiology, LMU University Hospital, LMU Munich, Munich, Germany
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
| | - Katharina Buerger
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy, Institute of Neuroscience and Physiology, University of Gothenburg, Mölndal, Gothenburg, Sweden
| | - Endy Weidinger
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Selim Guersel
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Schuster
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Anika Finze
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Stefanie Harris
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany
| | - Christian Wetzel
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Axel Rominger
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany
- Department of Nuclear Medicine, University of Bern, Inselspital, Bern, Switzerland
| | - Carla Palleis
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sabrina Katzdobler
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Lena Burow
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Carolin Kurz
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Mirlind Zaganjori
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Lena-Katharina Trappmann
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Oliver Goldhardt
- Department of Psychiatry and Psychotherapy, School of Medicine and Health, Technical University Munich, Klinikum Rechts Der Isar, Munich, Germany
| | - Timo Grimmer
- Department of Psychiatry and Psychotherapy, School of Medicine and Health, Technical University Munich, Klinikum Rechts Der Isar, Munich, Germany
| | - Jan Haeckert
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Daniel Keeser
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sophia Stoecklein
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | | | - Peter Bartenstein
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Günter U Höglinger
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Mikael Simons
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute of Neuronal Cell Biology, TU Munich, Munich, Germany
| | - Robert Perneczky
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
- Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, UK
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, University of Munich, Marchioninstraße 15, 81377, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
28
|
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
|
29
|
Hao N, Bai X, Hu A, Zhao G, Chen Y, Zhao J, Ling Q, Li X, Cai C, Wang Q, Wang Z, Fang J. Assessing the Global, Regional, and National Impact of High Body Mass Index on Alzheimer's Disease and Other Dementias Between 1990 and 2019. J Alzheimers Dis 2024; 97:293-307. [PMID: 38043013 DOI: 10.3233/jad-230827] [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: 12/04/2023]
Abstract
BACKGROUND Obesity significantly increases Alzheimer's disease (AD) and dementia risk. Understanding the link between a high body mass index (BMI) and these conditions is crucial for effective management and prevention. OBJECTIVE We aimed to estimate the burden of AD and other dementias attributed to high BMI from 1990 to 2019 based on sex, age, and socio-demographic indicators (SDI) at global, regional, and national levels. METHODS We collected data on deaths, disability-adjusted life years (DALYs), age-standardized mortality rates (ASMR), and age-standardized DALY rates (ASDR) from the 2019 Global Burden of Disease study for AD and dementia attributed to high BMI. We explored the correlation between SDI levels and ASDR. RESULTS In 2019, there were 198,476.2 deaths (95% UI: 32,695.4-593,366.4) and 3,159,912.4 DALYs (848,330.5-8,042,531) attributed to high BMI. Numbers of deaths, DALYs, ASMR, and ASDR increased since 1990. Females had higher deaths, ASMR, and ASDR than males. Mortality and DALYs rates increased with age. ASMR and ASDR increased across five SDI levels, with the highest rise in Low-middle SDI. High-income North America had the most deaths [30,993.9 (5,101.7-89,912.9)], while North Africa and the Middle East had the highest ASMR [4.61 (0.79-13.64)] and ASDR [72.56 (20.98-181.16)] in 2019. CONCLUSIONS The burden of AD and other dementias attributed to high BMI increased since 1990 globally and is still heaviest in developed regions. Females accounted predominantly for the burden than males. Timely measures are needed to against high BMI.
Collapse
Affiliation(s)
- Ning Hao
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xue Bai
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - An Hu
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Gaofeng Zhao
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yansheng Chen
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jianhe Zhao
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Qiong Ling
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiangyu Li
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Chuipu Cai
- College of Engineering, Shantou University, Shantou, China
| | - Qi Wang
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhaojun Wang
- Department of Thoracic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jiansong Fang
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
30
|
Perluigi M, Di Domenico F, Butterfield DA. Oxidative damage in neurodegeneration: roles in the pathogenesis and progression of Alzheimer disease. Physiol Rev 2024; 104:103-197. [PMID: 37843394 PMCID: PMC11281823 DOI: 10.1152/physrev.00030.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 03/30/2023] [Accepted: 05/24/2023] [Indexed: 10/17/2023] Open
Abstract
Alzheimer disease (AD) is associated with multiple etiologies and pathological mechanisms, among which oxidative stress (OS) appears as a major determinant. Intriguingly, OS arises in various pathways regulating brain functions, and it seems to link different hypotheses and mechanisms of AD neuropathology with high fidelity. The brain is particularly vulnerable to oxidative damage, mainly because of its unique lipid composition, resulting in an amplified cascade of redox reactions that target several cellular components/functions ultimately leading to neurodegeneration. The present review highlights the "OS hypothesis of AD," including amyloid beta-peptide-associated mechanisms, the role of lipid and protein oxidation unraveled by redox proteomics, and the antioxidant strategies that have been investigated to modulate the progression of AD. Collected studies from our groups and others have contributed to unraveling the close relationships between perturbation of redox homeostasis in the brain and AD neuropathology by elucidating redox-regulated events potentially involved in both the pathogenesis and progression of AD. However, the complexity of AD pathological mechanisms requires an in-depth understanding of several major intracellular pathways affecting redox homeostasis and relevant for brain functions. This understanding is crucial to developing pharmacological strategies targeting OS-mediated toxicity that may potentially contribute to slow AD progression as well as improve the quality of life of persons with this severe dementing disorder.
Collapse
Affiliation(s)
- Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi Fanelli," Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi Fanelli," Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States
| |
Collapse
|
31
|
Wang X, Sundermann EE, Buckley RF, Banks SJ. Sex differences in the association between tau PET and cognitive performance in a non-Hispanic White cohort with preclinical AD. Alzheimers Dement 2024; 20:25-33. [PMID: 37641484 PMCID: PMC10916995 DOI: 10.1002/alz.13432] [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: 03/03/2023] [Revised: 07/21/2023] [Accepted: 07/27/2023] [Indexed: 08/31/2023]
Abstract
INTRODUCTION We investigated how the associations between tau and cognitive measures differ by sex in the preclinical Alzheimer's disease (AD) stage. METHODS A total of 343 cognitively unimpaired, amyloid-positive individuals (205 women, 138 men) who self-identified as non-Hispanic White from the Anti-Amyloid Treatment in Asymptomatic Alzheimer's (A4) Study were included. We assessed sex-stratified associations between 18 F-flortaucipir positron emission tomography (PET) standardized uptake value ratio (SUVR) in the meta-temporal region and Preclinical Alzheimer's Cognitive Composite (PACC) and Computerized Cognitive Composite (C3) components. RESULTS We observed that higher tau level was significantly associated with worse cognitive performance only in women: PACC and its components except for Mini-Mental State Examination (MMSE) and C3 components: First Letter Name Recall (FNLT) and One-Card Learning Reaction Time (OCL RT). These associations except for FNLT were apolipoprotein E (APOE) ε4 independent. DISCUSSION Women show stronger associations between tau PET and cognitive outcomes in preclinical AD. These findings have important implications for sex-specific tau-targeted preventive AD clinical trials. HIGHLIGHTS The tau positron emission tomography (PET) signal in the meta-temporal region was associated with poor cognitive performance in preclinical Alzheimer's disease (AD). After sex stratification, the associations between regional tau PET and cognitive outcomes were observed only in women. The associations between tau PET and some cognitive outcomes were independent of apolipoprotein E (APOE) ε4.
Collapse
Affiliation(s)
- Xin Wang
- Department of NeurosciencesUniversity of CaliforniaSan DiegoCaliforniaUSA
| | | | - Rachel F. Buckley
- Department of NeurologyMassachusetts General Hospital/Harvard Medical SchoolBostonMassachusettsUSA
| | - Sarah J. Banks
- Department of NeurosciencesUniversity of CaliforniaSan DiegoCaliforniaUSA
- Department of PsychiatryUniversity of CaliforniaSan DiegoCaliforniaUSA
| | | |
Collapse
|
32
|
Wang D, Zheng J, Sun X, Xie L, Yang Y. Study on the Pharmacological Mechanism of Icariin for the Treatment of Alzheimer's Disease Based on Network Pharmacology and Molecular Docking Techniques. Metabolites 2023; 14:1. [PMID: 38276291 PMCID: PMC10820555 DOI: 10.3390/metabo14010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 01/27/2024] Open
Abstract
The purpose of this study is to explore the pharmacological mechanism of icariin (ICA) in the treatment of Alzheimer's disease (AD) based on network pharmacology and network molecular docking technology. In order to investigate the regulatory effect of ICA on the expression level of AD pathological phosphorylation regulatory proteins, this study further explored the possible molecular mechanism of ICA regulating AD autophagy through network pharmacology. Macromolecular docking network was verified by Autodock Vina 1.1.2 software. The main active ingredients of ICA, the physicochemical properties, and pharmacokinetic information of ICA were predicted using online databases and relevant information. The results showed that the targets of MAPK3, AKT1, HSP90AA1, ESR1, and HSP90AA1 were more critical in the treatment of AD. Autophagy, apoptosis, senescence factors, phosphatidylinositide 3-kinase/protein kinase B (P13K/AKT) signaling pathway, MAKP, mTOR, and other pathways were significantly associated with AD. Docking of ICA with HIF-1, BNIP3, PINK1, and Parkin pathway molecules showed that the key targets of the signaling pathway were more stably bound to ICA, which may provide a better pathway for ICA to regulate autophagy by providing a better pathway. ICA can improve AD, and its mechanism may be related to the P13K/AKT, MAKP, and mTOR signaling pathways, thereby regulating autophagy-related proteins.
Collapse
Affiliation(s)
- Dongwei Wang
- College of Police Dog Technology, Criminal Investigation Police University of China, Shenyang 110854, China; (D.W.); (J.Z.); (X.S.); (L.X.)
| | - Jilong Zheng
- College of Police Dog Technology, Criminal Investigation Police University of China, Shenyang 110854, China; (D.W.); (J.Z.); (X.S.); (L.X.)
| | - Xingsheng Sun
- College of Police Dog Technology, Criminal Investigation Police University of China, Shenyang 110854, China; (D.W.); (J.Z.); (X.S.); (L.X.)
| | - Liuwei Xie
- College of Police Dog Technology, Criminal Investigation Police University of China, Shenyang 110854, China; (D.W.); (J.Z.); (X.S.); (L.X.)
- The Second Affiliated Hospital of Shenyang Medical College, Shenyang 110031, China
| | - Yang Yang
- The Second Affiliated Hospital of Shenyang Medical College, Shenyang 110031, China
| |
Collapse
|
33
|
Zhou Z, Tarzanagh DA, Hou B, Tong B, Xu J, Feng Y, Long Q, Shen L. Fair Canonical Correlation Analysis. ADVANCES IN NEURAL INFORMATION PROCESSING SYSTEMS 2023; 36:3675-3705. [PMID: 38665178 PMCID: PMC11040228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
This paper investigates fairness and bias in Canonical Correlation Analysis (CCA), a widely used statistical technique for examining the relationship between two sets of variables. We present a framework that alleviates unfairness by minimizing the correlation disparity error associated with protected attributes. Our approach enables CCA to learn global projection matrices from all data points while ensuring that these matrices yield comparable correlation levels to group-specific projection matrices. Experimental evaluation on both synthetic and real-world datasets demonstrates the efficacy of our method in reducing correlation disparity error without compromising CCA accuracy.
Collapse
Affiliation(s)
| | | | | | | | - Jia Xu
- University of Pennsylvania
| | | | | | | |
Collapse
|
34
|
Blodgett JM, Ahmadi M, Stamatakis E, Rockwood K, Hamer M. Fractal complexity of daily physical activity and cognitive function in a midlife cohort. Sci Rep 2023; 13:20340. [PMID: 37990028 PMCID: PMC10663528 DOI: 10.1038/s41598-023-47200-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 11/10/2023] [Indexed: 11/23/2023] Open
Abstract
High stability of fluctuation in physiological patterns across fixed time periods suggest healthy fractal complexity, while greater randomness in fluctuation patterns may indicate underlying disease processes. The importance of fractal stability in mid-life remains unexplored. We quantified fractal regulation patterns in 24-h accelerometer data and examined associations with cognitive function in midlife. Data from 5097 individuals (aged 46) from the 1970 British Cohort Study were analyzed. Participants wore thigh-mounted accelerometers for seven days and completed cognitive tests (verbal fluency, memory, processing speed; derived composite z-score). Detrended fluctuation analysis (DFA) was used to examine temporal correlations of acceleration magnitude across 25 time scales (range: 1 min-10 h). Linear regression examined associations between DFA scaling exponents (DFAe) and each standardised cognitive outcome. DFAe was normally distributed (mean ± SD: 0.90 ± 0.06; range: 0.72-1.25). In males, a 0.10 increase in DFAe was associated with a 0.30 (95% Confidence Interval: 0.14, 0.47) increase in composite cognitive z-score in unadjusted models; associations were strongest for verbal fluency (0.10 [0.04, 0.16]). Associations remained in fully-adjusted models for verbal fluency only (0.06 [0.00, 0.12]). There was no association between DFA and cognition in females. Greater fractal stability in men was associated with better cognitive function. This could indicate mechanisms through which fractal complexity may scale up to and contribute to cognitive clinical endpoints.
Collapse
Affiliation(s)
- Joanna M Blodgett
- Institute of Sport Exercise and Health, Division of Surgery and Interventional Science, University College London, London, UK.
| | - Matthew Ahmadi
- School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Mackenzie Wearables Research Hub, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Emmanuel Stamatakis
- School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Mackenzie Wearables Research Hub, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Kenneth Rockwood
- Geriatric Medicine Research, Department of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Mark Hamer
- Institute of Sport Exercise and Health, Division of Surgery and Interventional Science, University College London, London, UK
| |
Collapse
|
35
|
Bolton CJ, Steinbach M, Khan OA, Liu D, O'Malley J, Dumitrescu L, Peterson A, Jefferson AL, Hohman TJ, Zetterberg H, Gifford KA. Clinical and demographic factors modify the association between plasma phosphorylated tau-181 and cognition. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.03.23298051. [PMID: 37961576 PMCID: PMC10635266 DOI: 10.1101/2023.11.03.23298051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
INTRODUCTION Plasma phosphorylated tau181 (p-tau181) associations with global cognition and memory are clear, but the link between p-tau181 with other cognitive domains and subjective cognitive decline (SCD) across the clinical spectrum of Alzheimer's disease (AD) and how this association changes based on genetic and demographic factors is poorly understood. METHODS Participants were drawn from the Alzheimer's Disease Neuroimaging Initiative and included 1185 adults aged >55 years with plasma p-tau181 and neuropsychological test data. Linear regression models related plasma p-tau181 to neuropsychological composite and SCD scores with follow-up models examining plasma p-tau181 interactions with cognitive diagnosis, APOE ε4 carrier status, age, and sex on cognitive outcomes. RESULTS Higher plasma p-tau181 was associated with worse memory, executive functioning, and language abilities, and greater informant-reported SCD. Visuospatial abilities and self-report SCD were not associated with plasma p-tau181. Associations were generally stronger in MCI or dementia, APOE ε4 carriers, women, and younger participants. DISCUSSION Higher levels of plasma p-tau181 are associated with worse neuropsychological test performance across multiple cognitive domains; however, these associations vary based on disease stage, genetic risk status, age, and sex.
Collapse
|
36
|
Balu D, Valencia-Olvera AC, Islam Z, Mielczarek C, Hansen A, Perez Ramos TM, York J, LaDu MJ, Tai LM. APOE genotype and sex modulate Alzheimer's disease pathology in aged EFAD transgenic mice. Front Aging Neurosci 2023; 15:1279343. [PMID: 38020764 PMCID: PMC10644540 DOI: 10.3389/fnagi.2023.1279343] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023] Open
Abstract
Increasing evidence supports that age, APOE and sex interact to modulate Alzheimer's disease (AD) risk, however the underlying pathways are unclear. One way that AD risk factors may modulate cognition is by impacting amyloid beta (Aβ) accumulation as plaques, and/or neuroinflammation Therefore, the goal of the present study was to evaluate the extent to which age, APOE and sex modulate Aβ pathology, neuroinflammation and behavior in vivo. To achieve this goal, we utilized the EFAD mice, which express human APOE3 or APOE4 and have five familial AD mutations (FAD) that result in Aβ42 overproduction. We assessed Aβ levels, reactive glia and Morris water maze performance in 6-, 10-, 14-, and 18-month-old EFAD mice. Female APOE4 mice had the highest Aβ deposition, fibrillar amyloid deposits and neuroinflammation as well as earlier behavior deficits. Interestingly, we found that female APOE3 mice and male APOE4 mice had similar levels of pathology. Collectively our data support that the combination of APOE4 and female sex is the most detrimental combination for AD, and that at older ages, female sex may be equivalent to APOE4 genotype.
Collapse
Affiliation(s)
- Deebika Balu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Ana C. Valencia-Olvera
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Zarak Islam
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
- University of Illinois College of Medicine, Chicago, IL, United States
| | - Clare Mielczarek
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Allison Hansen
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
- University of Illinois College of Medicine, Peoria, IL, United States
| | - Tamara M. Perez Ramos
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
- School of Medicine, St. George’s University, St. George’s, Grenada
| | - Jason York
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Leon M. Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
37
|
Park S, Lee S, Jeong KH. Predictors of Variation in the Cognitive Function Trajectories among Older Adults Living Alone: A Growth Mixture Modeling Approach. Healthcare (Basel) 2023; 11:2750. [PMID: 37893824 PMCID: PMC10606450 DOI: 10.3390/healthcare11202750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Considering the global aging population, this study investigates changes in cognitive function and predictive factors among older adults living alone. METHODS Using data collected from the Korean Longitudinal Study of Aging (KLoSA), the study examines 1217 participants to identify distinct cognitive change patterns and the variables affecting them. RESULTS Two primary cognitive function change types emerged: "High-Level Declining Type" and "Low-Level Stable Type." Although the former initially displayed normal cognitive function, it gradually declined over a period of 14 years until it reached mild cognitive impairment (MCI) levels by the year 2020. While the latter group had lower cognitive function from the beginning and remained stable throughout the study. Older age, female gender, rural residence, lower education, lower income, unemployment, and higher levels of depression were linked to a higher likelihood of belonging to the "Low-Level Stable Type". CONCLUSIONS The findings of these studies emphasize the need for proactive interventions and regular cognitive assessments for older individuals living alone, as cognitive impairment can develop even in individuals whose cognitive abilities are initially good. Also, tailored interventions should target specific demographic and socioeconomic groups to mitigate cognitive decline effectively.
Collapse
Affiliation(s)
- Soyoung Park
- Department of Social Welfare, Semyung University, 65 Semyung-ro, Jecheon 27136, Republic of Korea; (S.P.); (K.-H.J.)
| | - Seoyoon Lee
- Interdisciplinary Graduate Program in Social Welfare Policy, Yonsei University, 50 Yonsei-ro, Seoul 03722, Republic of Korea
| | - Kyu-Hyoung Jeong
- Department of Social Welfare, Semyung University, 65 Semyung-ro, Jecheon 27136, Republic of Korea; (S.P.); (K.-H.J.)
| |
Collapse
|
38
|
Haagsma AB, Souza DLB, Vasconcellos GM, Olandoski M, Jerez-Roig J, Baena CP. Longitudinal Relationship Between Handgrip Strength and Cognitive Function in a European Multicentric Population Older Than 50 Years. Phys Ther 2023; 103:pzad057. [PMID: 37249576 PMCID: PMC11009692 DOI: 10.1093/ptj/pzad057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/26/2022] [Accepted: 03/08/2023] [Indexed: 05/31/2023]
Abstract
OBJECTIVE The aim of this study was to analyze the bidirectional association between handgrip strength (HGS) and cognitive performance in different cognitive functions in a European population and to evaluate the predictive validity of HGS for the risk of future cognitive impairment in aging individuals. METHODS This was a prospective cohort study conducted using data on individuals over 50 years of age from the Survey of Health, Aging and Retirement in Europe (SHARE). HGS measures and scores in numeracy, recall, and verbal fluency were repeated and analyzed biannually for 4 years and were used in generalized estimating equations to test the bidirectional association, categorized by sex. RESULTS Of the 8236 individuals included, 55.73% were women with a mean age of 67.55 (standard deviation [SD] = 8.4) years and 44.27% were men with a mean age of 68.42 (SD = 7.7) years. HGS predicted cognitive decline in both sexes, except for numeracy in men, even after adjustments. The strongest association with HGS in women was in verbal fluency (β = .094; 95% CI = 0.039 to 0.151), whereas the strongest association with HGS in men was in delayed verbal recall (β = .095; 95% CI = 0.039 to 0.151). Conversely, the greatest cognitive predictor of HGS decline was verbal fluency in men (β = .796; 95% CI = 0.464 to 1.128), and in women (β = .801; 95% CI= 0.567 to 1.109). CONCLUSION There is a significant and bidirectional association between HGS and different cognitive functions in a European multicentric population. This bidirectional association differed between sexes. IMPACT Both men and women who presented with cognitive decline also showed early changes in their HGS measures, and vice versa, but there still were differences between the sexes. These findings reinforce that HGS may be a simple and inexpensive method to identify early signs of cognitive decline, and that studies and rehabilitation strategies should be more sex specific.
Collapse
Affiliation(s)
- Ariele B Haagsma
- Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Paraná, Brazil
| | - Dyego L B Souza
- Universidade Federal do Rio Grande do Norte (UFRN), Natal, Rio Grande do Norte, Brazil
| | | | - Márcia Olandoski
- Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Paraná, Brazil
| | - Javier Jerez-Roig
- Faculty of Health Sciences and Welfare, University of Vic - Central University of Catalonia (UVIC-UCC), Vic, Barcelona, Spain
| | - Cristina P Baena
- Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba, Paraná, Brazil
- Hospital Marcelino Champagnat, Curitiba, Paraná, Brazil
| |
Collapse
|
39
|
Pan F, Wang Y, Wang Y, Wang X, Guan Y, Xie F, Guo Q. Sex and APOE genotype differences in amyloid deposition and cognitive performance along the Alzheimer's Continuum. Neurobiol Aging 2023; 130:84-92. [PMID: 37481959 DOI: 10.1016/j.neurobiolaging.2023.06.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/25/2023]
Abstract
Conflicting findings exist regarding the differences in amyloid burden and cognitive performance based on sex and apolipoprotein E (APOE) genotype. This study aimed to investigate the brain amyloid-β (Aβ) burden and cognitive performances by sex and APOE genotype in a cohort of Aβ-positron emission tomography (PET)-positive participants. Brain Aβ burden was assessed using 18F-florbetapir PET standard uptake value ratios. Cognitive performance was evaluated using standardized neuropsychological tests. In the cognitively normal participants, females had a higher Aβ burden than males in APOE ε4 noncarriers, whereas APOE ε4 carriers had a higher Aβ burden than noncarriers in males. In the cognitively impaired participants, APOE ε4 carriers were more likely to have a higher Aβ burden than noncarriers in the brain regions of the lateral parietal gyrus, frontal gyrus, and precuneus. In addition, females were more likely to have poorer language and visuospatial performance compared to males, while the APOE genotype did not significantly impact cognitive performance. These findings further elucidate the impact of sex and APOE genotype on brain Aβ burden and sex-related cognitive performance should be considered in the Alzheimer's Continuum.
Collapse
Affiliation(s)
- Fengfeng Pan
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yifan Wang
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Wang
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoming Wang
- Department of Physiology, Capital Medical University, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Beijing, China
| | - Yihui Guan
- Department PET Center, Huashan Hospital, Fudan University, Shaznghai, China
| | - Fang Xie
- Department PET Center, Huashan Hospital, Fudan University, Shaznghai, China.
| | - Qihao Guo
- Department of Gerontology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
40
|
Huang J, Xu B, Chen X, Yang L, Liu D, Lin J, Liu Y, Lei X, Huang C, Dou W, Guo D, Wei X, Zhang P, Huang Y, Gu X, Zhang H. Sex Hormone-Binding Globulin and Risk of Incident Dementia in Middle-Aged to Older Women: Results from the UK Biobank Cohort Study. Neuroendocrinology 2023; 114:170-178. [PMID: 37725912 DOI: 10.1159/000533929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 08/28/2023] [Indexed: 09/21/2023]
Abstract
INTRODUCTION The association of serum sex hormone-binding globulin (SHBG) concentrations with dementia risk remains uncertain in middle-aged to older women. We examined associations of serum SHBG levels with incidence of all-cause dementia and its subtypes in middle-aged to older women from the large population-based UK Biobank cohort study. METHODS Serum total SHBG levels were measured by immunoassay. The incidence of all-cause dementia and its subtypes was recorded. Cox proportional hazards models were used to calculate hazard ratios (HR) for main outcomes. RESULTS Among 171,482 community-dwelling women (mean [SD] age was 59.9 [5.4] years, median follow-up of 11.8 years), 2,368 developed dementia, including 1,088 from Alzheimer's disease (AD), 451 from vascular dementia (VAD), and 1,609 from other dementia. After multivariable adjustments, higher serum SHBG levels were significantly associated with higher risks of all-cause dementia, AD, and other dementia (all p < 0.05). Compared to those in the lowest quartile of SHBG levels, participants in the highest quartile of SHBG levels had a higher risk of all-cause dementia (HR: 1.34; 95% confidence interval [CI]: 1.16-1.53), AD (HR: 1.32; 95% CI: 1.07-1.62), and other dementia (HR: 1.44; 95% CI: 1.21-1.70). However, this relationship was not significant for VAD (HR: 1.16; 95% CI: 0.86-1.56). CONCLUSION These findings indicated that higher serum SHBG concentrations were independently associated with higher risks of incident all-cause dementia, as well as AD and other dementia among middle-aged to older women. No association was found for VAD.
Collapse
Affiliation(s)
- Junlin Huang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bingyan Xu
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaomin Chen
- Department of Endocrinology, Zhongshan Hospital Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Linjie Yang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Deying Liu
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiayang Lin
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yating Liu
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xuzhen Lei
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chensihan Huang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weijuan Dou
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dan Guo
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xueyun Wei
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Peizhen Zhang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yan Huang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xuejiang Gu
- Dpartment of Endocrine and Metabolic Diseases, 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huijie Zhang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou, China
- State Key Laboratory of Organ Failure Research, Guangzhou, China
| |
Collapse
|
41
|
Scheffels JF, Ballasch I, Scheichel N, Voracek M, Kalbe E, Kessler J. The Influence of Age, Gender and Education on Neuropsychological Test Scores: Updated Clinical Norms for Five Widely Used Cognitive Assessments. J Clin Med 2023; 12:5170. [PMID: 37629212 PMCID: PMC10455991 DOI: 10.3390/jcm12165170] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/03/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Sociodemographic effects (i.e., age, gender, education) have been shown to influence neuropsychological test scores. The current retrospective, quasi-epidemiological work provides age-, gender- and education-corrected clinical norms for five common cognitive assessments. METHODS In total, test scores of 4968 patients from the University Hospital of Cologne (Department of Neurology), recruited between 2009 and 2020, were analyzed retrospectively. Conducted tests were the Mini-Mental State Examination (MMSE), F-A-S Test (FAS), Rey-Osterrieth Complex Figure Test (ROCFT) and Trail Making Test, Part A and B (TMT-A/-B). Using multiple linear regression analyses, test scores were analyzed for sociodemographic influences (age, gender, education). Based on these analyses, norms were generated by first separating patients into different age groups stratified by educational level and (if necessary) gender. Subsequently, percentile ranks and z-scores for a subsample including only individuals without dementia were calculated. RESULTS Lower age and higher educational level predicted better test scores (MMSE, FAS, ROCFT) and completion times (TMT-A/-B). Additionally, produced words on the FAS and remembered drawings from the ROCFT were influenced by gender, with females having better FAS but lower ROCFT (delayed recall) scores than males. Considering these effects, clinical norms were provided for the five cognitive assessments. CONCLUSIONS We found influences of age, gender and education on test scores, although they are frequently not or only partially considered for test score interpretation. With the provided norms, neuropsychologists can make more profound evaluations of cognitive performance. A user-friendly Microsoft Excel file is offered to assist this process.
Collapse
Affiliation(s)
| | - Isabell Ballasch
- Department of Medical Psychology, Neuropsychology & Gender Studies, Center for Neuropsychological Diagnostics and Intervention (CeNDI), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Nadine Scheichel
- Department of Medical Psychology, Neuropsychology & Gender Studies, Center for Neuropsychological Diagnostics and Intervention (CeNDI), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
- Department of Cognition, Emotion, and Methods in Psychology, Faculty of Psychology, University of Vienna, 1010 Vienna, Austria
| | - Martin Voracek
- Department of Cognition, Emotion, and Methods in Psychology, Faculty of Psychology, University of Vienna, 1010 Vienna, Austria
| | - Elke Kalbe
- Department of Medical Psychology, Neuropsychology & Gender Studies, Center for Neuropsychological Diagnostics and Intervention (CeNDI), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Josef Kessler
- Department of Medical Psychology, Neuropsychology & Gender Studies, Center for Neuropsychological Diagnostics and Intervention (CeNDI), Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| |
Collapse
|
42
|
Sims S, Barak O, Ryu V, Miyashita S, Kannangara H, Korkmaz F, Wizman S, Macdonald A, Gumerova A, Goosens K, Zaidi M, Yuen T, Lizneva D, Frolinger T. Absent LH signaling rescues the anxiety phenotype in aging female mice. Mol Psychiatry 2023; 28:3324-3331. [PMID: 37563278 DOI: 10.1038/s41380-023-02209-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/12/2023]
Abstract
Clinical studies and experimental data together support a role for pituitary gonadotropins, including luteinizing hormone (LH), otherwise considered solely as fertility hormones, in age-related cognitive decline. Furthermore, rising levels of LH in post-menopausal women have been implicated in the high prevalence of mood disorders. This study was designed to examine the effect of deficient LH signaling on both cognitive and emotional behavior in 12-month-old Lhcgr-/- mice. For this, we established and validated a battery of five tests, including Dark-Light Box (DLB), Y-Maze Spontaneous Alternation, Novel Object Recognition (NOR), and contextual and cued Fear Conditioning (FCT) tests. We found that 12-month-old female wild type mice display a prominent anxiety phenotype on DLB and FCT. This phenotype was not seen in 12-month-old female Lhcgr-/- mice, indicating full phenotypic rescue. Furthermore, there was no effect of LHCGR depletion on recognition memory or working spatial memory on NOR and Y-maze testing, respectively, in 12-month-old mice, notwithstanding the absence of a basal phenotype in wild type littermates. The latter data do not exclude an effect of LH on cognition documented in previous studies. Finally, 12-month-old male mice and 3-month-old male and female mice did not consistently display deficits on any test. The data collectively document, for the first time, that loss of LH signaling reverses age-related emotional disturbances, a prelude to future targeted therapies that block LH action.
Collapse
Affiliation(s)
- Steven Sims
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Orly Barak
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Vitaly Ryu
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sari Miyashita
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Hasni Kannangara
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Funda Korkmaz
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Soleil Wizman
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Anne Macdonald
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Anisa Gumerova
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ki Goosens
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Mone Zaidi
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Tony Yuen
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Daria Lizneva
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Tal Frolinger
- Center for Translational Medicine and Pharmacology, Departments of Pharmacological Sciences and of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
43
|
Digma LA, Litvan I, Del Ser T, Bayram E. Sex differences for cognitive decline in progressive supranuclear palsy. Parkinsonism Relat Disord 2023; 112:105454. [PMID: 37301014 PMCID: PMC10911684 DOI: 10.1016/j.parkreldis.2023.105454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/16/2023] [Accepted: 05/21/2023] [Indexed: 06/12/2023]
Abstract
INTRODUCTION Cognitive dysfunction is a core clinical feature of progressive supranuclear palsy (PSP), with executive function being most markedly affected. In other neurodegenerative conditions, such as Alzheimer's and Parkinson's diseases, there are a growing number of reports demonstrating that cognition is differentially impacted in men and women. In PSP, however, the sex differences in cognitive decline have yet to be fully characterized. METHODS Data were obtained from the TAUROS trial for 139 participants with mild-to-moderate PSP (62 women, 77 men). Sex differences in longitudinal change in cognitive performance were evaluated with linear mixed models. Exploratory subgroup analyses assessed whether sex differences varied by baseline executive dysfunction, PSP phenotype, or baseline age. RESULTS In the primary whole group analyses, there were no sex differences for change in cognitive performance. Among participants with normal executive function at baseline, men declined more severely on executive function and language tests. Among the PSP-Parkinsonism subgroup, men declined more severely on category fluency. Across people aged≥65, men had a worse decline on category fluency, whereas across people aged <65, women had a worse decline on DRS construction. CONCLUSION In people with mild-to-moderate PSP, there are no sex differences in cognitive decline. However, the rate of cognitive decline may differ for women and men based on the level of baseline executive dysfunction, PSP-phenotype and age. Further studies are needed to clarify how sex differences in PSP clinical progression vary by disease stage and to examine the contributions of co-pathology to these observed sex differences.
Collapse
Affiliation(s)
- Leonardino A Digma
- Parkinson and Other Movement Disorder Center, Department of Neurosciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| | - Irene Litvan
- Parkinson and Other Movement Disorder Center, Department of Neurosciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| | - Teodoro Del Ser
- Alzheimer's Disease Investigation Research Unit, CIEN Foundation, Carlos III Institute of Health, Queen Sofia Foundation, Alzheimer Research Centre, C. de Valderrebollo, 5, 28031, Madrid, Spain.
| | - Ece Bayram
- Parkinson and Other Movement Disorder Center, Department of Neurosciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| |
Collapse
|
44
|
Stanley M, Poupore N, Knisely K, Miller A, Imeh-Nathaniel A, Roley LT, Imeh-Nathaniel S, Goodwin R, Nathaniel TI. Differences in pharmacologic and demographic factors in male and female patients with vascular dementia, Alzheimer's disease, and mixed vascular dementia. FRONTIERS IN DEMENTIA 2023; 2:1137856. [PMID: 39081989 PMCID: PMC11285705 DOI: 10.3389/frdem.2023.1137856] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 06/15/2023] [Indexed: 08/02/2024]
Abstract
Background Increasing evidence suggests that demographic and pharmacologic factors may play a significant role in the epidemiology of dementia. Sex differences in prevalence also depend on dementia subtypes, such as Alzheimer's dementia (AD), vascular dementia (VaD), and mixed vascular-Alzheimer's dementia (MVAD). Therefore, studies are needed to investigate sex-specific differences, and identify potential therapeutic targets for both sexes. Methods Data was collected from the Prisma Health-Upstate Alzheimer's registry from 2016 to 2021 for 6,039 VaD patients, 9,290 AD patients, and 412 MVAD patients. A logistic regression was used to determine demographic and pharmacological factors associated with gender differences in patients with VaD, AD, and MVAD. Results In patients with VaD, African Americans (OR = 1.454, 95% CI, 1.257-1.682, p < 0.001) with increasing age (OR = 1.023, 95% CI, 1.017-1.029, p < 0.001), treated with aripiprazole (OR = 4.395, 95% CI, 2.880-6.707, p < 0.001), were associated with females, whereas patients treated with galantamine (OR = 0.228, 95% CI, 0.116-0.449, p < 0.001), memantine (OR = 0.662, 95% CI, 0.590-0.744, p < 0.001), with a history of tobacco (OR = 0.312, 95% CI, 0.278-0.349, p < 0.001), and ETOH (OR = 0.520, 95% CI, 0.452-0.598, p < 0.001) were associated with males. Among AD patients, African Americans (OR = 1.747, 95% CI, 1.486-2.053, p < 0.001), and Hispanics (OR = 3.668, 95% CI, 1.198-11.231, P = 0.023) treated with buspirone (OR = 1.541, 95% CI, 1.265-1.878, p < 0.001), and citalopram (OR = 1.790, 95% CI, 1.527-2.099, p < 0.001), were associated with females, whereas patients treated with memantine (OR = 0.882, 95% CI, 0.799-0.974, p = 0.013), and with a history of tobacco (OR = 0.247, 95% CI, 0.224-0.273, p < 0.001), and ETOH (OR = 0.627, 95% CI, 0.547-0.718, p < 0.001) were associated with male AD patients. In patients with MVAD, rivastigmine (OR = 3.293, 95% CI, 1.131-9.585, p = 0.029), memantine (OR = 2.816, 95% CI, 1.534-5.168, p < 0.001), and risperidone (OR = 10.515, 95% CI, 3.409-32.437, p < 0.001), were associated with females while patients with an increased length of stay (OR = 0.910, 95% CI, 0.828-1.000, p = 0.049), with a history of tobacco (OR = 0.148, 95% CI, 0.086-0.254, p < 0.001) and ETOH use (OR = 0.229, 95% CI, 0.110-0.477, p < 0.001) were more likely to be associated with males. Conclusions Our study revealed gender differences and similarities in the demographic and pharmacological factors of VaD, AD, and MVAD. Prospective studies are needed to determine the role of demographic and pharmacological factors in reducing sex-based disparities among VaD, AD, and MVAD patients.
Collapse
Affiliation(s)
- Madison Stanley
- School of Medicine Greenville, University of South Carolina, Greenville, SC, United States
| | - Nicolas Poupore
- School of Medicine Greenville, University of South Carolina, Greenville, SC, United States
| | - Krista Knisely
- School of Medicine Greenville, University of South Carolina, Greenville, SC, United States
| | - Alyssa Miller
- Department of Biology, North Greenville University, Tigerville, SC, United States
| | | | | | | | - Rich Goodwin
- School of Medicine Greenville, University of South Carolina, Greenville, SC, United States
| | - Thomas I. Nathaniel
- School of Medicine Greenville, University of South Carolina, Greenville, SC, United States
| |
Collapse
|
45
|
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
|
46
|
Liu M, Zhu L, Guo YJ, Zhang SS, Jiang L, Zhang Y, Chao FL, Tang Y. The effects of voluntary running exercise on the astrocytes of the medial prefrontal cortex in APP/PS1 mice. J Comp Neurol 2023. [PMID: 37146123 DOI: 10.1002/cne.25485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 03/07/2023] [Accepted: 03/20/2023] [Indexed: 05/07/2023]
Abstract
Pathological changes in the medial prefrontal cortex (mPFC) and astrocytes are closely associated with Alzheimer's disease (AD). Voluntary running has been found to effectively delay AD. However, the effects of voluntary running on mPFC astrocytes in AD are unclear. A total of 40 10-month-old male amyloid precursor protein/presenilin 1 (APP/PS1) mice and 40 wild-type (WT) mice were randomly divided into control and running groups, and the running groups underwent voluntary running for 3 months. Mouse cognition was assessed by the novel object recognition (NOR), Morris water maze (MWM), and Y maze tests. The effects of voluntary running on mPFC astrocytes were investigated using immunohistochemistry, immunofluorescence, western blotting, and stereology. APP/PS1 mice performed significantly worse than WT mice in the NOR, MWM, and Y maze tests, and voluntary running improved the performance of APP/PS1 mice in these tests. The total number of mPFC astrocytes was increased, cell bodies were enlarged, and protrusion number and length were increased in AD mice compared with WT mice, but there was no difference in component 3 (C3) levels in the mPFC (total mPFC level); however, C3 and S100B levels in astrocytes were increased in AD mice. Voluntary running reduced the total number of astrocytes and S100B levels in astrocytes and increased the density of PSD95+ puncta in direct contact with astrocyte protrusions in the APP/PS1 mouse mPFC. Three months of voluntary running inhibited astrocyte hyperplasia and S100B expression in astrocytes, increased the density of synapses in contact with astrocytes, and improved cognitive function in APP/PS1 mice.
Collapse
Affiliation(s)
- Mei Liu
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
- Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Lin Zhu
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
- Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Yi-Jing Guo
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
- Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Shan-Shan Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Army Medical University, Chongqing, P. R. China
| | - Lin Jiang
- Laboratory Teaching & Management Center, Chongqing Medical University, Chongqing, P. R. China
| | - Yi Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, P. R. China
| | - Feng-Lei Chao
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
- Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| | - Yong Tang
- Department of Histology and Embryology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
- Laboratory of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, P. R. China
| |
Collapse
|
47
|
Polsinelli AJ, Logan PE, Lane KA, Manchella MK, Nemes S, Sanjay AB, Gao S, Apostolova LG. APOE ε4 carrier status and sex differentiate rates of cognitive decline in early- and late-onset Alzheimer's disease. Alzheimers Dement 2023; 19:1983-1993. [PMID: 36394443 PMCID: PMC10182251 DOI: 10.1002/alz.12831] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 07/19/2022] [Accepted: 09/19/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND We studied the effect of apolipoprotein E (APOE) ε4 status and sex on rates of cognitive decline in early- (EO) and late- (LO) onset Alzheimer's disease (AD). METHOD We ran mixed-effects models with longitudinal cognitive measures as dependent variables, and sex, APOE ε4 carrier status, and interaction terms as predictor variables in 998 EOAD and 2562 LOAD participants from the National Alzheimer's Coordinating Center. RESULTS APOE ε4 carriers showed accelerated cognitive decline relative to non-carriers in both EOAD and LOAD, although the patterns of specific cognitive domains that were affected differed. Female participants showed accelerated cognitive decline relative to male participants in EOAD only. The effect of APOE ε4 was greater in EOAD for executive functioning (p < 0.0001) and greater in LOAD for language (p < 0.0001). CONCLUSION We found APOE ε4 effects on cognitive decline in both EOAD and LOAD and female sex in EOAD only. The specific patterns and magnitude of decline are distinct between the two disease variants. HIGHLIGHTS Apolipoprotein E (APOE) ε4 carrier status and sex differentiate rates of cognitive decline in early-onset (EO) and late-onset (LO) Alzheimer's disease (AD). APOE ε4 in EOAD accelerated decline in memory, executive, and processing speed domains. Female sex in EOAD accelerated decline in language, memory, and global cognition. The effect of APOE ε4 was stronger for language in LOAD and for executive function in EOAD. Sex effects on language and executive function decline differed between EOAD and LOAD.
Collapse
Affiliation(s)
- Angelina J. Polsinelli
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Alzheimer’s Disease Research Center, Indianapolis, Indiana, USA
| | - Paige E. Logan
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Alzheimer’s Disease Research Center, Indianapolis, Indiana, USA
| | - Kathleen A. Lane
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Mohit K. Manchella
- Department of Chemistry, University of Southern Indiana Evansville, Indiana, USA
| | - Sára Nemes
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | - Sujuan Gao
- Indiana Alzheimer’s Disease Research Center, Indianapolis, Indiana, USA
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Liana G. Apostolova
- Department of Neurology, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Indiana Alzheimer’s Disease Research Center, Indianapolis, Indiana, USA
| |
Collapse
|
48
|
Iulita MF, Bejanin A, Vilaplana E, Carmona-Iragui M, Benejam B, Videla L, Barroeta I, Fernández S, Altuna M, Pegueroles J, Montal V, Valldeneu S, Giménez S, González-Ortiz S, Torres S, El Bounasri El Bennadi S, Padilla C, Rozalem Aranha M, Estellés T, Illán-Gala I, Belbin O, Valle-Tamayo N, Camacho V, Blessing E, Osorio RS, Videla S, Lehmann S, Holland AJ, Zetterberg H, Blennow K, Alcolea D, Clarimón J, Zaman SH, Blesa R, Lleó A, Fortea J. Association of biological sex with clinical outcomes and biomarkers of Alzheimer's disease in adults with Down syndrome. Brain Commun 2023; 5:fcad074. [PMID: 37056479 PMCID: PMC10088472 DOI: 10.1093/braincomms/fcad074] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 12/07/2022] [Accepted: 03/16/2023] [Indexed: 03/19/2023] Open
Abstract
The study of sex differences in Alzheimer's disease is increasingly recognized as a key priority in research and clinical development. People with Down syndrome represent the largest population with a genetic link to Alzheimer's disease (>90% in the 7th decade). Yet, sex differences in Alzheimer's disease manifestations have not been fully investigated in these individuals, who are key candidates for preventive clinical trials. In this double-centre, cross-sectional study of 628 adults with Down syndrome [46% female, 44.4 (34.6; 50.7) years], we compared Alzheimer's disease prevalence, as well as cognitive outcomes and AT(N) biomarkers across age and sex. Participants were recruited from a population-based health plan in Barcelona, Spain, and from a convenience sample recruited via services for people with intellectual disabilities in England and Scotland. They underwent assessment with the Cambridge Cognitive Examination for Older Adults with Down Syndrome, modified cued recall test and determinations of brain amyloidosis (CSF amyloid-β 42 / 40 and amyloid-PET), tau pathology (CSF and plasma phosphorylated-tau181) and neurodegeneration biomarkers (CSF and plasma neurofilament light, total-tau, fluorodeoxyglucose-PET and MRI). We used within-group locally estimated scatterplot smoothing models to compare the trajectory of biomarker changes with age in females versus males, as well as by apolipoprotein ɛ4 carriership. Our work revealed similar prevalence, age at diagnosis and Cambridge Cognitive Examination for Older Adults with Down Syndrome scores by sex, but males showed lower modified cued recall test scores from age 45 compared with females. AT(N) biomarkers were comparable in males and females. When considering apolipoprotein ɛ4, female ɛ4 carriers showed a 3-year earlier age at diagnosis compared with female non-carriers (50.5 versus 53.2 years, P = 0.01). This difference was not seen in males (52.2 versus 52.5 years, P = 0.76). Our exploratory analyses considering sex, apolipoprotein ɛ4 and biomarkers showed that female ɛ4 carriers tended to exhibit lower CSF amyloid-β 42/amyloid-β 40 ratios and lower hippocampal volume compared with females without this allele, in line with the clinical difference. This work showed that biological sex did not influence clinical and biomarker profiles of Alzheimer's disease in adults with Down syndrome. Consideration of apolipoprotein ɛ4 haplotype, particularly in females, may be important for clinical research and clinical trials that consider this population. Accounting for, reporting and publishing sex-stratified data, even when no sex differences are found, is central to helping advance precision medicine.
Collapse
Affiliation(s)
- M Florencia Iulita
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona 08025, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid 28031, Spain
- Women’s Brain Project, Guntershausen 8357, Switzerland
| | - Alexandre Bejanin
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona 08025, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid 28031, Spain
| | - Eduard Vilaplana
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona 08025, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid 28031, Spain
| | - Maria Carmona-Iragui
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona 08025, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid 28031, Spain
- Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona 08029, Spain
| | - Bessy Benejam
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid 28031, Spain
- Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona 08029, Spain
| | - Laura Videla
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona 08025, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid 28031, Spain
- Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona 08029, Spain
| | - Isabel Barroeta
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona 08025, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid 28031, Spain
| | - Susana Fernández
- Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona 08029, Spain
| | - Miren Altuna
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona 08025, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid 28031, Spain
| | - Jordi Pegueroles
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona 08025, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid 28031, Spain
| | - Victor Montal
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona 08025, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid 28031, Spain
| | - Silvia Valldeneu
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona 08025, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid 28031, Spain
| | - Sandra Giménez
- Multidisciplinary Sleep Unit, Hospital de la Santa Creu i Sant Pau, Barcelona 08041, Spain
| | | | - Soraya Torres
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona 08025, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid 28031, Spain
| | - Shaimaa El Bounasri El Bennadi
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona 08025, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid 28031, Spain
| | - Concepcion Padilla
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona 08025, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid 28031, Spain
| | - Mateus Rozalem Aranha
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona 08025, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid 28031, Spain
| | - Teresa Estellés
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona 08025, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid 28031, Spain
| | - Ignacio Illán-Gala
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona 08025, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid 28031, Spain
| | - Olivia Belbin
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona 08025, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid 28031, Spain
| | - Natalia Valle-Tamayo
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona 08025, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid 28031, Spain
| | - Valle Camacho
- Nuclear Medicine Department, Hospital de la Santa Creu i Sant Pau, Barcelona 08041, Spain
| | - Esther Blessing
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Ricardo S Osorio
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Sebastian Videla
- Clinical Research Support Unit, Bellvitge Biomedical Research Institute (IDIBELL), Department of Clinical Pharmacology, University of Barcelona, Barcelona 08908, Spain
| | - Sylvain Lehmann
- Institute for Neurosciences of Montpellier, Institute for Regenerative Medicine and Biotherapy, Université de Montpellier, CHU de Montpellier, INSERM, Montpellier 34295, France
| | - Anthony J Holland
- Department of Psychiatry, Cambridge Intellectual and Developmental Disabilities Research Group, University of Cambridge, Douglas House, Cambridge CB2 8AH, United Kingdom
- Cambridgeshire & Peterborough NHS Foundation Trust, Fulbourn Hospital, Cambridge CB21 5EF, United Kingdom
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Möndal 40530, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal 40530, Sweden
- UK Dementia Research Institute, University College London, London WC1E 6BT, United Kingdom
- Department of Neurodegenerative Disease, University College London Institute of Neurology, London WC1E 6BT, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong 1512-1518, China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Möndal 40530, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal 40530, Sweden
| | - Daniel Alcolea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona 08025, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid 28031, Spain
| | - Jordi Clarimón
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona 08025, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid 28031, Spain
| | - Shahid H Zaman
- Department of Psychiatry, Cambridge Intellectual and Developmental Disabilities Research Group, University of Cambridge, Douglas House, Cambridge CB2 8AH, United Kingdom
- Cambridgeshire & Peterborough NHS Foundation Trust, Fulbourn Hospital, Cambridge CB21 5EF, United Kingdom
| | - Rafael Blesa
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona 08025, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid 28031, Spain
| | - Alberto Lleó
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona 08025, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid 28031, Spain
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona 08025, Spain
- Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid 28031, Spain
- Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona 08029, Spain
| |
Collapse
|
49
|
Sánchez CQ, Schmitt FW, Curdt N, Westhoff AC, Bänfer IWH, Bayer TA, Bouter Y. Search Strategy Analysis of 5xFAD Alzheimer Mice in the Morris Water Maze Reveals Sex- and Age-Specific Spatial Navigation Deficits. Biomedicines 2023; 11:biomedicines11020599. [PMID: 36831135 PMCID: PMC9953202 DOI: 10.3390/biomedicines11020599] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/03/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
Spatial disorientation and navigational impairments are not only some of the first memory deficits in Alzheimer's disease, but are also very disease-specific. In rodents, the Morris Water Maze is used to investigate spatial navigation and memory. Here, we examined the spatial memory in the commonly used 5xFAD Alzheimer mouse model in a sex- and age-dependent manner. Our findings show first spatial learning deficits in 7-month-old female 5xFAD and 12-month-old male 5xFAD mice, respectively. While the assessment of spatial working memory using escape latencies provides a global picture of memory performance, it does not explain how an animal solves a spatial task. Therefore, a detailed analysis of swimming strategies was performed to better understand the behavioral differences between 5xFAD and WT mice. 5xFAD mice used a qualitatively and quantitatively different search strategy pattern than wildtype animals that used more non-spatial strategies and showed allocentric-specific memory deficits. Furthermore, a detailed analysis of swimming strategies revealed allocentric memory deficits in the probe trial in female 3-month-old and male 7-month-old 5xFAD animals before the onset of severe reference memory deficits. Overall, we could demonstrate that spatial navigation deficits in 5xFAD mice are age- and sex-dependent, with female mice being more severely affected. In addition, the implementation of a search strategy classification system allowed an earlier detection of behavioral differences and therefore could be a powerful tool for preclinical drug testing in the 5xFAD model.
Collapse
Affiliation(s)
- Carolina Quintanilla Sánchez
- Department of Psychiatry and Psychotherapy, Division of Molecular Psychiatry, University Medical Center (UMG), Georg-August-University, 37075 Goettingen, Germany
| | - Franziska W. Schmitt
- Department of Psychiatry and Psychotherapy, Division of Molecular Psychiatry, University Medical Center (UMG), Georg-August-University, 37075 Goettingen, Germany
| | - Nadine Curdt
- Department of Psychiatry and Psychotherapy, Division of Molecular Psychiatry, University Medical Center (UMG), Georg-August-University, 37075 Goettingen, Germany
| | - Anna Celine Westhoff
- Department of Psychiatry and Psychotherapy, Division of Molecular Psychiatry, University Medical Center (UMG), Georg-August-University, 37075 Goettingen, Germany
| | - Irina Wanda Helene Bänfer
- Department of Psychiatry and Psychotherapy, Division of Molecular Psychiatry, University Medical Center (UMG), Georg-August-University, 37075 Goettingen, Germany
| | - Thomas A. Bayer
- Department of Psychiatry and Psychotherapy, Division of Molecular Psychiatry, University Medical Center (UMG), Georg-August-University, 37075 Goettingen, Germany
| | - Yvonne Bouter
- Department of Psychiatry and Psychotherapy, Division of Molecular Psychiatry, University Medical Center (UMG), Georg-August-University, 37075 Goettingen, Germany
- Department of Nuclear Medicine, University Medical Center Göttingen (UMG), 37075 Goettingen, Germany
- Correspondence:
| |
Collapse
|
50
|
Saleh RNM, Hornberger M, Ritchie CW, Minihane AM. Hormone replacement therapy is associated with improved cognition and larger brain volumes in at-risk APOE4 women: results from the European Prevention of Alzheimer's Disease (EPAD) cohort. Alzheimers Res Ther 2023; 15:10. [PMID: 36624497 PMCID: PMC9830747 DOI: 10.1186/s13195-022-01121-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 11/08/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND The risk of dementia is higher in women than men. The metabolic consequences of estrogen decline during menopause accelerate neuropathology in women. The use of hormone replacement therapy (HRT) in the prevention of cognitive decline has shown conflicting results. Here we investigate the modulating role of APOE genotype and age at HRT initiation on the heterogeneity in cognitive response to HRT. METHODS The analysis used baseline data from participants in the European Prevention of Alzheimer's Dementia (EPAD) cohort (total n= 1906, women= 1178, 61.8%). Analysis of covariate (ANCOVA) models were employed to test the independent and interactive impact of APOE genotype and HRT on select cognitive tests, such as MMSE, RBANS, dot counting, Four Mountain Test (FMT), and the supermarket trolley test (SMT), together with volumes of the medial temporal lobe (MTL) regions by MRI. Multiple linear regression models were used to examine the impact of age of HRT initiation according to APOE4 carrier status on these cognitive and MRI outcomes. RESULTS APOE4 HRT users had the highest RBANS delayed memory index score (P-APOE*HRT interaction = 0.009) compared to APOE4 non-users and to non-APOE4 carriers, with 6-10% larger entorhinal (left) and amygdala (right and left) volumes (P-interaction= 0.002, 0.003, and 0.005 respectively). Earlier introduction of HRT was associated with larger right (standardized β= -0.555, p=0.035) and left hippocampal volumes (standardized β= -0.577, p=0.028) only in APOE4 carriers. CONCLUSION HRT introduction is associated with improved delayed memory and larger entorhinal and amygdala volumes in APOE4 carriers only. This may represent an effective targeted strategy to mitigate the higher life-time risk of AD in this large at-risk population subgroup. Confirmation of findings in a fit for purpose RCT with prospective recruitment based on APOE genotype is needed to establish causality.
Collapse
Affiliation(s)
- Rasha N M Saleh
- Norwich Medical School, University of East Anglia, Norwich, UK.
| | | | - Craig W Ritchie
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | | |
Collapse
|