Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Age Ageing ; 53(Suppl 2): ii47-ii59, 2024 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-38745492

RESUMO

Hippocampal neurogenesis (HN) occurs throughout the life course and is important for memory and mood. Declining with age, HN plays a pivotal role in cognitive decline (CD), dementia, and late-life depression, such that altered HN could represent a neurobiological susceptibility to these conditions. Pertinently, dietary patterns (e.g., Mediterranean diet) and/or individual nutrients (e.g., vitamin D, omega 3) can modify HN, but also modify risk for CD, dementia, and depression. Therefore, the interaction between diet/nutrition and HN may alter risk trajectories for these ageing-related brain conditions. Using a subsample (n = 371) of the Three-City cohort-where older adults provided information on diet and blood biobanking at baseline and were assessed for CD, dementia, and depressive symptomatology across 12 years-we tested for interactions between food consumption, nutrient intake, and nutritional biomarker concentrations and neurogenesis-centred susceptibility status (defined by baseline readouts of hippocampal progenitor cell integrity, cell death, and differentiation) on CD, Alzheimer's disease (AD), vascular and other dementias (VoD), and depressive symptomatology, using multivariable-adjusted logistic regression models. Increased plasma lycopene concentrations (OR [95% CI] = 1.07 [1.01, 1.14]), higher red meat (OR [95% CI] = 1.10 [1.03, 1.19]), and lower poultry consumption (OR [95% CI] = 0.93 [0.87, 0.99]) were associated with an increased risk for AD in individuals with a neurogenesis-centred susceptibility. Increased vitamin D consumption (OR [95% CI] = 1.05 [1.01, 1.11]) and plasma γ-tocopherol concentrations (OR [95% CI] = 1.08 [1.01, 1.18]) were associated with increased risk for VoD and depressive symptomatology, respectively, but only in susceptible individuals. This research highlights an important role for diet/nutrition in modifying dementia and depression risk in individuals with a neurogenesis-centred susceptibility.


Assuntos
Disfunção Cognitiva , Demência , Depressão , Hipocampo , Neurogênese , Estado Nutricional , Humanos , Idoso , Masculino , Feminino , Depressão/psicologia , Depressão/metabolismo , Depressão/sangue , Disfunção Cognitiva/sangue , Disfunção Cognitiva/psicologia , Disfunção Cognitiva/epidemiologia , Demência/psicologia , Demência/epidemiologia , Demência/sangue , Demência/etiologia , Fatores de Risco , Hipocampo/metabolismo , Envelhecimento/psicologia , Idoso de 80 Anos ou mais , Cognição , Fatores Etários , Dieta/efeitos adversos , Envelhecimento Cognitivo/psicologia , Biomarcadores/sangue
2.
Mol Nutr Food Res ; 65(23): e2100606, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34661340

RESUMO

SCOPE: Diet is considered an important modulator of cognitive decline and dementia, but the available evidence is, however, still fragmented and often inconsistent. METHODS AND RESULTS: The article studies the long-term prospective Three-City Cohort, which consists of two separate nested case-control sample sets from different geographic regions (Bordeaux, n = 418; Dijon, n = 424). Cognitive decline is evaluated through five neuropsychological tests (Mini-Mental State Examination, Benton Visual Retention Test, Isaac's Set Test, Trail-Making Test part A, and Trail-Making Test part B). The food-related and microbiota-derived circulating metabolome is studied in participants free of dementia at baseline, by subjecting serum samples to large-scale quantitative metabolomics analysis. A protective association is found between metabolites derived from cocoa, coffee, mushrooms, red wine, the microbial metabolism of polyphenol-rich foods, and cognitive decline, as well as a negative association with metabolites related to unhealthy dietary components, such as artificial sweeteners and alcohol. CONCLUSION: These results provide insight into the early metabolic events that are associated with the later risk to develop cognitive decline within the crosstalk between diet, gut microbiota and the endogenous metabolism, which can help identify potential targets for preventive and therapeutic strategies to preserve cognitive health.


Assuntos
Disfunção Cognitiva , Microbioma Gastrointestinal , Microbiota , Idoso , Disfunção Cognitiva/etiologia , Alimentos , Humanos , Estudos Prospectivos
3.
Nutrients ; 13(2)2021 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-33498469

RESUMO

Early-life stress (ES) exposure increases the risk of developing obesity. Breastfeeding can markedly decrease this risk, and it is thought that the physical properties of the lipid droplets in human milk contribute to this benefit. A concept infant milk formula (IMF) has been developed that mimics these physical properties of human milk (Nuturis®, N-IMF). Previously, we have shown that N-IMF reduces, while ES increases, western-style diet (WSD)-induced fat accumulation in mice. Peripheral and central inflammation are considered to be important for obesity development. We therefore set out to test the effects of ES, Nuturis® and WSD on adipose tissue inflammatory gene expression and microglia in the arcuate nucleus of the hypothalamus. ES was induced in mice by limiting the nesting and bedding material from postnatal day (P) 2 to P9. Mice were fed a standard IMF (S-IMF) or N-IMF from P16 to P42, followed by a standard diet (STD) or WSD until P230. ES modulated adipose tissue inflammatory gene expression early in life, while N-IMF had lasting effects into adulthood. Centrally, ES led to a higher microglia density and more amoeboid microglia at P9. In adulthood, WSD increased the number of amoeboid microglia, and while ES exposure increased microglia coverage, Nuturis® reduced the numbers of amoeboid microglia upon the WSD challenge. These results highlight the impact of the early environment on central and peripheral inflammatory profiles, which may be key in the vulnerability to develop metabolic derangements later in life.


Assuntos
Dieta Ocidental , Fórmulas Infantis , Inflamação , Microglia , Animais , Feminino , Masculino , Tecido Adiposo/metabolismo , Animais Recém-Nascidos , Contagem de Células , Citocinas/metabolismo , Hipotálamo/citologia , Inflamação/etiologia , Inflamação/prevenção & controle , Macrófagos/fisiologia , Camundongos Endogâmicos BALB C , Microglia/citologia , Estresse Psicológico , Camundongos
4.
Sci Transl Med ; 10(448)2018 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-29973407

RESUMO

Dementia is a major social and economic problem for our aging population. One of the most common of dementia in the elderly is cerebral small vessel disease (SVD). Magnetic resonance scans of SVD patients typically show white matter abnormalities, but we do not understand the mechanistic pathological link between blood vessels and white matter myelin damage. Hypertension is suggested as the cause of sporadic SVD, but a recent alternative hypothesis invokes dysfunction of the blood-brain barrier as the primary cause. In a rat model of SVD, we show that endothelial cell (EC) dysfunction is the first change in development of the disease. Dysfunctional ECs secrete heat shock protein 90α, which blocks oligodendroglial differentiation, contributing to impaired myelination. Treatment with EC-stabilizing drugs reversed these EC and oligodendroglial pathologies in the rat model. EC and oligodendroglial dysfunction were also observed in humans with early, asymptomatic SVD pathology. We identified a loss-of-function mutation in ATPase11B, which caused the EC dysfunction in the rat SVD model, and a single-nucleotide polymorphism in ATPase11B that was associated with white matter abnormalities in humans with SVD. We show that EC dysfunction is a cause of SVD white matter vulnerability and provide a therapeutic strategy to treat and reverse SVD in the rat model, which may also be of relevance to human SVD.


Assuntos
Doenças de Pequenos Vasos Cerebrais/patologia , Doenças de Pequenos Vasos Cerebrais/fisiopatologia , Endotélio Vascular/fisiopatologia , Substância Branca/patologia , Adenosina Trifosfatases/genética , Animais , Barreira Hematoencefálica/patologia , Barreira Hematoencefálica/fisiopatologia , Proliferação de Células , Modelos Animais de Doenças , Células Endoteliais/patologia , Endotélio Vascular/patologia , Proteínas de Choque Térmico HSP90/metabolismo , Homozigoto , Humanos , Hipertensão/patologia , Hipertensão/fisiopatologia , Proteínas de Membrana Transportadoras/genética , Pessoa de Meia-Idade , Células Precursoras de Oligodendrócitos/metabolismo , Polimorfismo de Nucleotídeo Único/genética , Ratos , Substância Branca/fisiopatologia
5.
Neurobiol Dis ; 103: 163-173, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28400135

RESUMO

Alzheimer's disease (AD) is a highly prevalent multifactorial disease for which Diabetes Mellitus (DM) is a risk factor. Abnormal phosphorylation and aggregation of tau is a key hallmark of AD. In animal models, DM induces or exacerbates the phosphorylation of tau, suggesting that DM may influence the risk at AD by directly facilitating tau pathology. Previously we reported that tau phosphorylation induced in response to metabolic stress is reversible. Since identification and understanding of early players in tau pathology is pivotal for therapeutic intervention, we here investigated the mechanism underlying tau phosphorylation in the diabetic brain and its potential for reversibility. To model DM we used streptozotocin-treatment to induce insulin deficiency in rats. Insulin depletion leads to increased tau phosphorylation in the brain and we investigated the activation status of known tau kinases and phosphatases in this model. We identified protein kinase A (PKA) as a tau kinase activated by DM in the brain. The potential relevance of this signaling pathway to AD pathogenesis is indicated by the increased level of active PKA in temporal cortex of early stage AD patients. Our data indicate that activation of PKA and tau phosphorylation are associated with insulin deficiency per se, rather than the downstream energy deprivation. In vitro studies confirm that insulin deficiency results in PKA activation and tau phosphorylation. Strikingly, both active PKA and induced tau phosphorylation are reversed upon insulin treatment in the steptozotocin animal model. Our data identify insulin deficiency as a direct trigger that induces the activity of the tau kinase PKA and results in tau phosphorylation. The reversibility upon insulin treatment underscores the potential of insulin as an early disease-modifying intervention in AD and other tauopathies.


Assuntos
Encéfalo/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Insulina/deficiência , Proteínas tau/metabolismo , Idoso , Idoso de 80 Anos ou mais , Animais , Encéfalo/patologia , Linhagem Celular Tumoral , Ativação Enzimática/fisiologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fosforilação/fisiologia , Ratos , Ratos Wistar
6.
Brain Behav Immun ; 63: 160-175, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28027926

RESUMO

Exposure to stress during the sensitive period of early-life increases the risk to develop cognitive impairments and psychopathology later in life. In addition, early-life stress (ES) exposure, next to genetic causes, has been proposed to modulate the development and progression of Alzheimer's disease (AD), however evidence for this hypothesis is currently lacking. We here tested whether ES modulates progression of AD-related neuropathology and assessed the possible contribution of neuroinflammatory factors in this. We subjected wild-type (WT) and transgenic APP/PS1 mice, as a model for amyloid neuropathology, to chronic ES from postnatal day (P)2 to P9. We next studied how ES exposure affected; 1) amyloid ß (Aß) pathology at an early (4month old) and at a more advanced pathological (10month old) stage, 2) neuroinflammatory mediators immediately after ES exposure as well as in adult WT mice, and 3) the neuroinflammatory response in relation to Aß neuropathology. ES exposure resulted in a reduction of cell-associated amyloid in 4month old APP/PS1 mice, but in an exacerbation of Aß plaque load at 10months of age, demonstrating that ES affects Aß load in the hippocampus in an age-dependent manner. Interestingly, ES modulated various neuroinflammatory mediators in the hippocampus of WT mice as well as in response to Aß neuropathology. In WT mice, immediately following ES exposure (P9), Iba1-immunopositive microglia exhibited reduced complexity and hippocampal interleukin (IL)-1ß expression was increased. In contrast, microglial Iba1 and CD68 were increased and hippocampal IL-6 expression was decreased at 4months, while these changes resolved by 10months of age. Finally, Aß neuropathology triggered a neuroinflammatory response in APP/PS1 mice that was altered after ES exposure. APP/PS1 mice exhibited increased CD68 expression at 4months, which was further enhanced by ES, whereas the microglial response to Aß neuropathology, as measured by Iba1 and CD11b, was less prominent after ES at 10months of age. Finally, the hippocampus appears to be more vulnerable for these ES-induced effects, since ES did not affect Aß neuropathology and neuroinflammation in the entorhinal cortex of adult ES exposed mice. Overall, our results demonstrate that ES exposure has both immediate and lasting effects on the neuroinflammatory response. In the context of AD, such alterations in neuroinflammation might contribute to aggravated neuropathology in ES exposed mice, hence altering disease progression. This indicates that, at least in a genetic context, ES could aggravate AD pathology.


Assuntos
Doença de Alzheimer/patologia , Amiloide/fisiologia , Estresse Psicológico/imunologia , Doença de Alzheimer/metabolismo , Amiloide/imunologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Amiloidose/patologia , Animais , Modelos Animais de Doenças , Progressão da Doença , Hipocampo/metabolismo , Humanos , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Camundongos , Camundongos Transgênicos , Microglia/metabolismo , Neuroimunomodulação/imunologia , Placa Amiloide , Estresse Psicológico/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA