Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 657
Filter
1.
Neuroimage ; 301: 120881, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39362507

ABSTRACT

White matter (WM) development has been studied extensively, but most studies used cross-sectional data, and to the best of our knowledge, none of them considered the possible effects of biological (vs. chronological) age. Therefore, we conducted a longitudinal multimodal study of WM development and studied changes in fractional anisotropy (FA) in the different WM tracts and their relationship with cortical thickness-based measures of brain aging in young adulthood. A total of 105 participants from the European Longitudinal Study of Pregnancy and Childhood (ELSPAC) prenatal birth cohort underwent magnetic resonance imaging (MRI) at the age of 23-24, and the age of 28-30 years. At both time points, FA in the different WM tracts was extracted using the JHU atlas, and brain age gap estimate (BrainAGE) was calculated using the Neuroanatomical Age Prediction using R (NAPR) model based on cortical thickness maps. Changes in FA and the speed of cortical brain aging were calculated as the difference between the respective variables in the late vs. early 20s. We demonstrated tract-specific increases as well as decreases in FA, which indicate that the WM microstructure continues to develop in the third decade of life. Moreover, the significant interaction between the speed of cortical brain aging, tract, and sex on mean FA revealed that a greater speed of cortical brain aging in young adulthood predicted greater decreases in FA in the bilateral cingulum and left superior longitudinal fasciculus in young adult men. Overall, these changes in FA in the WM tracts in young adulthood point out the protracted development of WM microstructure, particularly in men.

2.
Hum Brain Mapp ; 45(13): e70005, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39225381

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.


Subject(s)
Aging , Brain , Deep Learning , Magnetic Resonance Imaging , Sex Characteristics , Humans , Female , Male , Middle Aged , Aged , Aging/physiology , Brain/physiology , Brain/diagnostic imaging , Connectome/methods , Nerve Net/physiology , Nerve Net/diagnostic imaging
3.
Trends Neurosci ; 47(10): 751-752, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39237428

ABSTRACT

Neuroinflammation is a feature of both neurodegenerative disease and normal brain aging. The roles of type I interferon (IFN-I) in the aged brain are incompletely understood. A recent article by Roy et al. reveals pervasive IFN-I activity in normal mouse brain aging, and highlights the importance of microglial IFN-I signaling in neuroinflammation.


Subject(s)
Aging , Interferon Type I , Neuroinflammatory Diseases , Animals , Interferon Type I/metabolism , Interferon Type I/immunology , Aging/immunology , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/metabolism , Humans , Brain/metabolism , Brain/immunology , Microglia/metabolism , Microglia/immunology , Signal Transduction/physiology , Mice
4.
Int Immunopharmacol ; 142(Pt B): 113151, 2024 Dec 05.
Article in English | MEDLINE | ID: mdl-39303538

ABSTRACT

The current study aimed to evaluate the preventive effects of urolithin C (Uro C), a gut microbial metabolite of ellagitannins on D-galactose (D-gal)-induced brain damage during the aging process and to elucidate the underlying mechanisms. In our study, the protective effect of Uro C on D-gal-induced BV2 microglia cell-mediated neuroinflammation damage in primary cortical neurons in vitro was confirmed. The results in an aging model in vivo induced by D-gal demonstrated that Uro C prevented D-gal-induced memory impairment, long-term potentiation (LTP) damage, and synaptic dysfunction through behavioral, electrophysiological, and histological examinations. Additionally, amyloidogenesis was observed in the central nervous system. The findings indicated that Uro C exhibited a preventive effect on the D-gal-induced elevation of ß-amyloid (1-42 specific) (Aß1-42) accumulation, APP levels, ABCE1 levels, and the equilibrium of the cholinergic system in the aging mouse brain. Moreover, Uro C demonstrated downregulation of D-gal-induced glial overactivation through inhibition of the MAPK/NF-kB pathway. This resulted in the regulation of inflammatory mediators and cytokines, including iNOS, IL-6, IL-1ß, and TNF-ɑ, in the mouse brain and BV2 microglial cells. Taken together, our results suggested that Uro C treatment could effectively mitigate the D-gal-induced memory impairment and amyloidogenesis, and the underlying mechanism might be tightly related to the improvement of neuroinflammation by suppressing the MAPK/NF-kB pathway, indicating Uro C might be an alternative and promising agent for the treatment of aging and age-associated brain diseases.


Subject(s)
Aging , Amyloid beta-Peptides , Cognitive Dysfunction , Coumarins , Galactose , Hydrolyzable Tannins , Microglia , NF-kappa B , Signal Transduction , Animals , Coumarins/pharmacology , Coumarins/therapeutic use , NF-kappa B/metabolism , Hydrolyzable Tannins/pharmacology , Hydrolyzable Tannins/therapeutic use , Aging/drug effects , Aging/metabolism , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Mice , Amyloid beta-Peptides/metabolism , Male , Microglia/drug effects , Microglia/metabolism , Signal Transduction/drug effects , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Down-Regulation/drug effects , Cell Line , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Brain/drug effects , Brain/metabolism , Brain/pathology , Cells, Cultured
5.
Sci Bull (Beijing) ; 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39332926

ABSTRACT

Age-related cognitive decline and dementia are significant manifestations of brain aging. As the elderly population grows rapidly, the health and socio-economic impacts of cognitive dysfunction have become increasingly significant. Although clinical treatment of dementia has faced considerable challenges over the past few decades, with limited breakthroughs in slowing its progression, there has been substantial progress in understanding the molecular mechanisms and hallmarks of age-related dementia (ARD). This progress brings new hope for the intervention and treatment of this disease. In this review, we categorize the latest findings in ARD biomarkers into four stages based on disease progression: healthy brain, pre-clinical, mild cognitive impairment, and dementia. We then systematically summarize the most promising therapeutic approaches to prevent or slow ARD at four levels: genome and epigenome, organelle, cell, and organ and organism. We emphasize the importance of early prevention and detection, along with the implementation of combined treatments as multimodal intervention strategies, to address brain aging and ARD in the future.

6.
Geroscience ; 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39312153

ABSTRACT

The brain of higher organisms, such as nonhuman primates, is particularly rich in lipids, with a gray to white matter ratio of approximately 40 to 60%. White matter primarily consists of lipids, and during normal aging, it undergoes significant degeneration due to myelin pathology, which includes structural abnormalities, like sheath splitting, and local inflammation. Cognitive decline in normal aging, without neurodegenerative diseases, is strongly linked to myelin pathology. Although the exact cause of myelin damage is unclear, older myelin differs from younger myelin, as shown by electron microscopy and altered expression of myelin-related RNAs. However, changes in lipid composition during brain aging remain poorly understood. This study assessed lipid profiles from the frontal lobe corpus callosum, an area where age-related myelin pathology is linked to cognitive decline. Results showed significant changes in lipids with age, revealing distinct age-related profiles. Some lipids that are enriched in myelin sheaths become more saturated, while important structural components, like ceramides, decrease. Disease-associated biomarkers such as cholesterol ester Che (22:6) and sulfatide ST (42:2) also change in older monkeys. Additionally, gene expression of lipid biosynthetic enzymes declines with age, while lipid peroxidation remains stable in the same brain region. This suggests that changes in lipid biosynthesis, rather than oxidative damage, likely account for the differences in lipid composition. Our findings indicate that myelin in the normal aging monkey brain shows diverse lipid changes, which may relate to age-related myelin pathology and could constitute targets for designing nutrient supplements or drugs to rejuvenate the brain's lipidome.

7.
Neurooncol Adv ; 6(1): vdae118, 2024.
Article in English | MEDLINE | ID: mdl-39220249

ABSTRACT

Background: The incidence of brain cancer and neurodegenerative diseases is increasing with a demographic shift towards aging populations. Biological parallels have been observed between glioblastoma and Alzheimer's disease (AD), which converge on accelerated brain aging. Here, we aimed to map the cooccurrence of AD neuropathological change (ADNC) in the tumor-adjacent cortex of patients with glioblastoma. Methods: Immunohistochemical screening of AD markers amyloid beta (Abeta), amyloid precursor protein (APP), and hyperphosphorylated tau (pTau) was conducted in 420 tumor samples of 205 patients. For each cortex area, we quantified ADNC, neurons, tumor cells, and microglia. Results: Fifty-two percent of patients (N = 106/205) showed ADNC (Abeta and pTau, Abeta or pTau) in the tumor-adjacent cortex, with histological patterns widely consistent with AD. ADNC was positively correlated with patient age and varied spatially according to Thal phases and Braak stages. It decreased with increasing tumor cell infiltration (P < .0001) and was independent of frequent expression of APP in neuronal cell bodies (N = 182/205) and in tumor necrosis-related axonal spheroids (N = 195/205; P = .46). Microglia response was most present in tumor cell infiltration plus ADNC, being further modulated by patient age and sex. ADNC did not impact patient survival in the present cohort. Conclusions: Our findings highlight the frequent presence of ADNC in the glioblastoma vicinity, which was linked to patient age and tumor location. The cooccurrence of AD and glioblastoma seemed stochastic without clear spatial relation. ADNC did not impact patient survival in our cohort.

8.
Politics Life Sci ; : 1-13, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39291658

ABSTRACT

The cognitive deterioration of politicians is a critical emerging issue. As professions including law and medicine develop and implement cognitive assessments, their insights may inform the proper strategy within politics. The aging, lifetime-appointed judiciary raises legal and administrative questions of such assessments, while testing of older physicians experiencing cognitive decline provides real-life examples of implementation. In politics, cognitive assessment must contend with the field's unique challenges, also taking context-dependent interpretations of cognitive-neuropsychological status into account. These perspectives, from legal and medical experts, political scientists, and officeholders, can contribute toward an equitable, functioning, and non-discriminatory system of assessing cognition that educates the public and enables politicians to maintain their public responsibilities. With proper implementation and sufficient public knowledge, we believe cognitive assessments for politicians, particularly political candidates, can be valuable for maintaining properly functioning governance. We offer recommendations on the development, implementation, and execution of such assessments, grappling with their democratic and legal implications.

9.
Biogerontology ; 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39300009

ABSTRACT

Oxidative stress-induced neuronal apoptosis is primarily involved in brain aging and impaired hippocampal neurogenesis. Long-term D-galactose administration increases oxidative stress related to brain aging. Chrysin, a subtype of flavonoids, exhibits neuroprotective effects, particularly its antioxidant properties. To elucidate the neuroprotection of chrysin on neuronal apoptosis and an impaired hippocampal neurogenesis relevant to oxidative damage in D-galactose-induced brain aging, male Sprague Dawley rats were allocated into vehicle control, D-galactose, chrysin, and cotreated rats. The rats received their respective treatments daily for 8 weeks. The reactions of scavenging enzymes, protein regulating endogenous antioxidant defense, and anti-apoptotic protein expression were significantly reduced in the hippocampus and prefrontal cortex of the animals receiving D-galactose. Conversely, product of oxidative damage and apoptotic protein expressions were significantly elevated in both cortical areas of the D-galactose group. In hippocampal neurogenesis, significant upregulation of cell cycle arrest and decrease in differentiated protein expression were detected after D-galactose administration. Nevertheless, chrysin supplementation significantly mitigated all negative effects in animals receiving D-galactose. This study demonstrates that chrysin likely attenuates brain aging induced by D-galactose by enhancing scavenging enzyme activities and reducing oxidative stress, neuronal apoptosis, and the impaired hippocampal neurogenesis.

10.
Eur J Pharmacol ; 983: 176959, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39216746

ABSTRACT

Aging is an inevitable law of the process of life during which many physiological functions change. Brain aging is an important mechanism in the occurrence and development of degenerative diseases of the central nervous system. ß-Hydroxybutyrate (BHBA) is a water-soluble, endogenous small-molecule ketone that can cross the blood-brain barrier and induce neuroprotective effects. This study aimed to investigate the effects of BHBA on D-galactose (D-gal) induced aging in mice and its underlying mechanisms using in vitro and in vivo experiments. These results indicated that D-gal-induced senescence, oxidative stress, and inflammatory responses were inhibited by BHBA, and autophagy was promoted by BHBA. Mechanistically, we explored the role of metastasis-associated antigen-1 (MTA1) in D-gal-induced damaged in HT22 cells using small interfering RNA (siRNA). The results demonstrated that the expression of MTA1 was significantly increased by BHBA, which attenuated D-gal-induced aging, oxidative stress, and inflammatory responses, and promoted autophagy through the upregulation of MTA1. In conclusion, MTA1 may be a novel target for treating aging caused by neurological damage. BHBA improves brain aging by activating the MTA1 pathway.


Subject(s)
3-Hydroxybutyric Acid , Aging , Autophagy , Brain , Galactose , Oxidative Stress , Signal Transduction , Animals , 3-Hydroxybutyric Acid/pharmacology , 3-Hydroxybutyric Acid/therapeutic use , Mice , Aging/drug effects , Oxidative Stress/drug effects , Male , Autophagy/drug effects , Brain/drug effects , Brain/metabolism , Brain/pathology , Signal Transduction/drug effects , Cell Line , Repressor Proteins/metabolism , Repressor Proteins/genetics , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Mice, Inbred C57BL , Trans-Activators/metabolism , Trans-Activators/genetics
11.
Neuroimage ; 299: 120825, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39214438

ABSTRACT

As an important biomarker of neural aging, the brain age reflects the integrity and health of the human brain. Accurate prediction of brain age could help to understand the underlying mechanism of neural aging. In this study, a cross-stratified ensemble learning algorithm with staking strategy was proposed to obtain brain age and the derived predicted age difference (PAD) using T1-weighted magnetic resonance imaging (MRI) data. The approach was characterized as by implementing two modules: one was three base learners of 3D-DenseNet, 3D-ResNeXt, 3D-Inception-v4; another was 14 secondary learners of liner regressions. To evaluate performance, our method was compared with single base learners, regular ensemble learning algorithms, and state-of-the-art (SOTA) methods. The results demonstrated that our proposed model outperformed others models, with three metrics of mean absolute error (MAE), root mean-squared error (RMSE), and coefficient of determination (R2) of 2.9405 years, 3.9458 years, and 0.9597, respectively. Furthermore, there existed significant differences in PAD among the three groups of normal control (NC), mild cognitive impairment (MCI) and Alzheimer's disease (AD), with an increased trend across NC, MCI, and AD. It was concluded that the proposed algorithm could be effectively used in computing brain aging and PAD, and offering potential for early diagnosis and assessment of normal brain aging and AD.


Subject(s)
Aging , Alzheimer Disease , Brain , Cognitive Dysfunction , Magnetic Resonance Imaging , Humans , Brain/diagnostic imaging , Aging/physiology , Aged , Magnetic Resonance Imaging/methods , Male , Female , Alzheimer Disease/diagnostic imaging , Cognitive Dysfunction/diagnostic imaging , Aged, 80 and over , Middle Aged , Machine Learning , Algorithms
12.
Int J Mol Sci ; 25(15)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39125687

ABSTRACT

Human brain aging is characterized by the production and deposition of ß-amyloid (Aß) in the form of senile plaques and cerebral amyloid angiopathy and the intracellular accumulation of hyper-phosphorylated tau (Hp-tau) to form neurofibrillary tangles (NFTs) and dystrophic neurites of senile plaques. The process progresses for years and eventually manifests as cognitive impairment and dementia in a subgroup of aged individuals. Aß is produced and deposited first in the neocortex in most aged mammals, including humans; it is usually not accompanied by altered behavior and cognitive impairment. Hp-tau is less frequent than Aß pathology, and NFTs are rare in most mammals. In contrast, NFTs are familiar from middle age onward in humans; NFTs first appear in the paleocortex and selected brain stem nuclei. NFTs precede for decades or years Aß deposition and correlate with dementia in about 5% of individuals at the age of 65 and 25% at the age of 85. Based on these comparative data, (a) Aß deposition is the most common Alzheimer's disease neuropathological change (ADNC) in the brain of aged mammals; (b) Hp-tau is less common, and NFTs are rare in most aged mammals; however, NFTs are the principal cytoskeletal pathology in aged humans; (c) NFT in aged humans starts in selected nuclei of the brain stem and paleocortical brain regions progressing to the most parts of the neocortex and other regions of the telencephalon; (d) human brain aging is unique among mammalian species due to the early appearance and dramatic progression of NFTs from middle age onward, matching with cognitive impairment and dementia in advanced cases; (e) neither mammalian nor human brain aging supports the concept of the amyloid cascade hypothesis.


Subject(s)
Aging , Alzheimer Disease , Neurofibrillary Tangles , tau Proteins , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Animals , Humans , Aging/pathology , Aging/metabolism , Neurofibrillary Tangles/pathology , Neurofibrillary Tangles/metabolism , tau Proteins/metabolism , Brain/pathology , Brain/metabolism , Amyloid beta-Peptides/metabolism , Mammals/metabolism , Plaque, Amyloid/pathology , Plaque, Amyloid/metabolism
13.
Alzheimers Dement (Amst) ; 16(3): e70001, 2024.
Article in English | MEDLINE | ID: mdl-39183745

ABSTRACT

INTRODUCTION: We examined the relationship between sedentary behavior (SB), moderate-to-vigorous physical activity (MVPA), and white matter hyperintensity (WMH) volumes, a common magnetic resonance imaging (MRI) marker associated with risk of neurodegenerative disease in middle-aged to older adults. METHODS: We used data from the UK Biobank (n = 14,415; 45 to 81 years) that included accelerometer-derived measures of SB and MVPA, and WMH volumes from MRI. RESULTS: Both MVPA and SB were associated with WMH volumes (ßMVPA = -0.03 [-0.04, -0.01], p < 0.001; ßSB = 0.02 [0.01, 0.03], p = 0.007). There was a significant interaction between SB and MVPA on WMH volumes (ßSB×MVPA = -0.015 [-0.028, -0.001], p SB×MVPA = 0.03) where SB was positively associated with WMHs at low MVPA, and MVPA was negatively associated with WMHs at high SB. DISCUSSION: While this study cannot establish causality, the results highlight the potential importance of considering both MVPA and SB in strategies aimed at reducing the accumulation of WMH volumes in middle-aged to older adults. Highlights: SB is associated with greater WMH volumes and MVPA is associated with lower WMH volumes.Relationships between SB and WMH are strongest at low levels of MVPA.Associations between MVPA and WMH are strongest at high levels of SB.Considering both SB and MVPA may be effective strategies for reducing WMHs.

14.
Front Aging Neurosci ; 16: 1429005, 2024.
Article in English | MEDLINE | ID: mdl-39149145

ABSTRACT

The cGAS-STING pathway is a pivotal element of the innate immune system, recognizing cytosolic DNA to initiate the production of type I interferons and pro-inflammatory cytokines. This study investigates the alterations of the cGAS-STING signaling components in the cortex and hippocampus of mice aged 24 and 108 weeks. In the cortex of old mice, an increase in the dsDNA sensor protein cGAS and its product 2'3'-cGAMP was observed, without corresponding activation of downstream signaling, suggesting an uncoupling of cGAS activity from STING activation. This phenomenon may be attributed to increased dsDNA concentrations in the EC neurons, potentially arising from nuclear DNA damage. Contrastingly, the hippocampus did not exhibit increased cGAS activity with aging, but there was a notable elevation in STING levels, particularly in microglia, neurons and astrocytes. This increase in STING did not correlate with enhanced IRF3 activation, indicating that brain inflammation induced by the cGAS-STING pathway may manifest extremely late in the aging process. Furthermore, we highlight the role of autophagy and its interplay with the cGAS-STING pathway, with evidence of autophagy dysfunction in aged hippocampal neurons leading to STING accumulation. These findings underscore the complexity of the cGAS-STING pathway's involvement in brain aging, with regional variations in activity and potential implications for neurodegenerative diseases.

15.
Bioengineering (Basel) ; 11(7)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-39061729

ABSTRACT

The intricate dynamics of brain aging, especially the neurodegenerative mechanisms driving accelerated (ABA) and resilient brain aging (RBA), are pivotal in neuroscience. Understanding the temporal dynamics of these phenotypes is crucial for identifying vulnerabilities to cognitive decline and neurodegenerative diseases. Currently, there is a lack of comprehensive understanding of the temporal dynamics and neuroimaging biomarkers linked to ABA and RBA. This study addressed this gap by utilizing a large-scale UK Biobank (UKB) cohort, with the aim to elucidate brain aging heterogeneity and establish the foundation for targeted interventions. Employing Lasso regression on multimodal neuroimaging data, structural MRI (sMRI), diffusion MRI (dMRI), and resting-state functional MRI (rsfMRI), we predicted the brain age and classified individuals into ABA and RBA cohorts. Our findings identified 1949 subjects (6.2%) as representative of the ABA subpopulation and 3203 subjects (10.1%) as representative of the RBA subpopulation. Additionally, the Discriminative Event-Based Model (DEBM) was applied to estimate the sequence of biomarker changes across aging trajectories. Our analysis unveiled distinct central ordering patterns between the ABA and RBA cohorts, with profound implications for understanding cognitive decline and vulnerability to neurodegenerative disorders. Specifically, the ABA cohort exhibited early degeneration in four functional networks and two cognitive domains, with cortical thinning initially observed in the right hemisphere, followed by the temporal lobe. In contrast, the RBA cohort demonstrated initial degeneration in the three functional networks, with cortical thinning predominantly in the left hemisphere and white matter microstructural degeneration occurring at more advanced stages. The detailed aging progression timeline constructed through our DEBM analysis positioned subjects according to their estimated stage of aging, offering a nuanced view of the aging brain's alterations. This study holds promise for the development of targeted interventions aimed at mitigating age-related cognitive decline.

16.
Geroscience ; 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39014131

ABSTRACT

Frailty reflects age-related damage to multiple physiological systems. Executive dysfunction is often a presenting symptom of diseases characterized by cognitive impairment. A decline in cardiovascular health is associated with worse executive function. We tested the hypothesis that higher frailty would be associated with executive dysfunction and that cardiovascular health would mediate this relationship. Middle- and older-aged adults at baseline (n = 29,591 [51% female]) and 3-year follow-up (n = 25,488 [49% females]) from the Canadian Longitudinal Study on Aging (comprehensive cohort) were included. Frailty was determined at baseline from a 61-item index, a cumulative cardiovascular health score was calculated from 30 variables at baseline, and participants completed a word-color Stroop task as an assessment of executive function. Multiple linear regressions and mediation analyses of cardiovascular health were conducted between frailty, Stroop interference-condition reaction time, and cardiovascular health in groups stratified by both age and sex (middle-aged males [MM], middle-aged females [MF], older-aged males [OM], older-aged females [OF]). Frailty (MM, 0.15 ± 0.05; MF, 0.16 ± 0.06; OM, 0.21 ± 0.06; OF, 0.23 ± 0.06) was negatively associated with cardiovascular health (MM, 0.12 ± 0.08; MF, 0.11 ± 0.07; OM, 0.20 ± 0.10; OF, 0.18 ± 0.09; ß > 0.037, p < 0.001), as well as the Stroop reaction time at 3-year follow-up (MM, 23.7 ± 7.9; MF, 23.1 ± 7.3; OM, 32.9 ± 13.1; OF, 30.9 ± 12.0; ß > 2.57, p < 0.001) across all groups when adjusted for covariates. Cardiovascular health was a partial (~ 10%) mediator between frailty and reaction time, aside from MFs. In conclusion, higher frailty levels are associated with executive dysfunction, which was partially mediated by cardiovascular health. Strategies to improve frailty and better cardiovascular health may be useful for combatting the age-related decline in executive function.

17.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39000227

ABSTRACT

The notion of notable anatomical, biochemical, and behavioral distinctions within male and female brains has been a contentious topic of interest within the scientific community over several decades. Advancements in neuroimaging and molecular biological techniques have increasingly elucidated common mechanisms characterizing brain aging while also revealing disparities between sexes in these processes. Variations in cognitive functions; susceptibility to and progression of neurodegenerative conditions, notably Alzheimer's and Parkinson's diseases; and notable disparities in life expectancy between sexes, underscore the significance of evaluating aging within the framework of gender differences. This comprehensive review surveys contemporary literature on the restructuring of brain structures and fundamental processes unfolding in the aging brain at cellular and molecular levels, with a focus on gender distinctions. Additionally, the review delves into age-related cognitive alterations, exploring factors influencing the acceleration or deceleration of aging, with particular attention to estrogen's hormonal support of the central nervous system.


Subject(s)
Aging , Brain , Humans , Brain/diagnostic imaging , Brain/metabolism , Brain/physiology , Aging/physiology , Female , Sex Characteristics , Male , Animals , Cognition/physiology , Sex Factors
18.
Article in English | MEDLINE | ID: mdl-39033955

ABSTRACT

BACKGROUND: Brain aging is a complex process that involves functional alterations in multiple subnetworks and brain regions. However, most previous studies investigating aging-related functional connectivity (FC) changes using resting-state functional magnetic resonance images (rs-fMRIs) have primarily focused on the linear correlation between brain subnetworks, ignoring the nonlinear casual properties of fMRI signals. METHODS: We introduced the neural Granger causality technique to investigate the sex-dependent nonlinear Granger connectivity (NGC) during aging on a publicly available dataset of 227 healthy participants acquired cross-sectionally in Leipzig, Germany. RESULTS: Our findings indicate that brain aging may cause widespread declines in NGC at both regional and subnetwork scales. These findings exhibit high reproducibility across different network sparsities, demonstrating the efficacy of static and dynamic analysis strategies. Females exhibit greater heterogeneity and reduced stability in NGC compared to males during aging, especially the NGC between the visual network and other subnetworks. Besides, NGC strengths can well reflect the individual cognitive function, which may therefore work as a sensitive metric in cognition-related experiments for individual-scale or group-scale mechanism understanding. CONCLUSION: These findings indicate that NGC analysis is a potent tool for identifying sex-dependent brain aging patterns. Our results offer valuable perspectives that could substantially enhance the understanding of sex differences in neurological diseases in the future, especially in degenerative disorders.


Subject(s)
Aging , Brain , Magnetic Resonance Imaging , Sex Characteristics , Humans , Male , Female , Brain/physiology , Brain/diagnostic imaging , Aging/physiology , Middle Aged , Aged , Adult , Young Adult , Cross-Sectional Studies , Nonlinear Dynamics , Nerve Net/diagnostic imaging , Nerve Net/physiology , Neural Pathways/physiology , Neural Pathways/diagnostic imaging , Aged, 80 and over
19.
J Alzheimers Dis ; 100(s1): S153-S164, 2024.
Article in English | MEDLINE | ID: mdl-39031364

ABSTRACT

Senile plaques, mainly diffuse, and cerebral amyloid-ß (Aß) angiopathy are prevalent in the aging brain of non-human primates, from lemurs to non-human Hominidae. Aß but not hyper-phosphorylated tau (HPtau) pathology is the common nominator proteinopathy of non-human primate brain aging. The abundance of Aß in the aging primate brain is well tolerated, and the impact on cognitive functions is usually limited to particular tasks. In contrast, human brain aging is characterized by the early appearance of HPtau pathology, mainly forming neurofibrillary tangles, dystrophic neurites of neuritic plaques, and neuropil threads, preceding Aß deposits by several decades and by its severity progressing from selected nuclei of the brain stem, entorhinal cortex, and hippocampus to the limbic system, neocortex, and other brain regions. Neurofibrillary tangles correlate with cognitive impairment and dementia in advanced cases. Aß pathology is linked in humans to altered membrane protein and lipid composition, particularly involving lipid rafts. Although similar membrane alterations are unknown in non-human primates, membrane senescence is postulated to cause the activated ß-amyloidogenic pathway, and Aß pathology is the prevailing signature of non-human and human primate brain aging.


Subject(s)
Aging , Amyloid beta-Peptides , Brain , Primates , Animals , Aging/pathology , Aging/metabolism , Brain/pathology , Brain/metabolism , Amyloid beta-Peptides/metabolism , Humans , Neurofibrillary Tangles/pathology , Plaque, Amyloid/pathology , tau Proteins/metabolism
20.
J Neurochem ; 168(9): 2243-2263, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38973579

ABSTRACT

Myelin water fraction (MWF) imaging has emerged as a promising magnetic resonance imaging (MRI) biomarker for investigating brain function and composition. This comprehensive review synthesizes the current state of knowledge on MWF as a biomarker of human cerebral aging, neurodegenerative diseases, and risk factors influencing myelination. The databases used include Web of Science, Scopus, Science Direct, and PubMed. We begin with a brief discussion of the theoretical foundations of MWF imaging, including its basis in MR physics and the mathematical modeling underlying its calculation, with an overview of the most adopted MRI methods of MWF imaging. Next, we delve into the clinical and research applications that have been explored to date, highlighting its advantages and limitations. Finally, we explore the potential of MWF to serve as a predictive biomarker for neurological disorders and identify future research directions for optimizing MWF imaging protocols and interpreting MWF in various contexts. By harnessing the power of MWF imaging, we may gain new insights into brain health and disease across the human lifespan, ultimately informing novel diagnostic and therapeutic strategies.


Subject(s)
Aging , Biomarkers , Magnetic Resonance Imaging , Myelin Sheath , Neurodegenerative Diseases , Humans , Myelin Sheath/metabolism , Neurodegenerative Diseases/diagnostic imaging , Neurodegenerative Diseases/metabolism , Aging/metabolism , Biomarkers/metabolism , Risk Factors , Magnetic Resonance Imaging/methods , Brain/diagnostic imaging , Brain/metabolism , Water/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL