Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 8 de 8
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
iScience ; 26(7): 106996, 2023 Jul 21.
Article de Anglais | MEDLINE | ID: mdl-37534143

RÉSUMÉ

The structure and function of the cardiovascular system are modulated across the day by circadian rhythms, making this system susceptible to circadian rhythm disruption. Recent evidence demonstrated that short-term exposure to a pervasive circadian rhythm disruptor, artificial light at night (ALAN), increased inflammation and altered angiogenic transcripts in the hippocampi of mice. Here, we examined the effects of four nights of ALAN exposure on mouse hippocampal vascular networks. To do this, we analyzed 2D and 3D images of hippocampal vasculature and hippocampal transcriptomic profiles of mice exposed to ALAN. ALAN reduced vascular density in the CA1 and CA2/3 of female mice and the dentate gyrus of male mice. Network structure and connectivity were also impaired in the CA2/3 of female mice. These results demonstrate the rapid and potent effects of ALAN on cerebrovascular networks, highlighting the importance of ALAN mitigation in the context of health and cerebrovascular disease.

2.
Neurobiol Aging ; 105: 115-128, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34062487

RÉSUMÉ

Heart disease and vascular disease positively correlate with the incidence of Alzheimer's disease (AD). Although there is ostensible involvement of dysfunctional cerebrovasculature in AD pathophysiology, the characterization of the specific changes and development of vascular injury during AD remains unclear. In the present study, we established a time-course for the structural changes and degeneration of the angioarchitecture in AD. We used cerebrovascular corrosion cast and µCT imaging to evaluate the geometry, topology, and complexity of the angioarchitecture in the brain of wild type and 3xTg AD mice. We hypothesized that changes to the microvasculature occur early during the disease, and these early identifiable aberrations would be more prominent in the brain subregions implicated in the cognitive decline of AD. Whole-brain analysis of the angioarchitecture indicated early morphological abnormalities and degeneration of microvascular networks in 3xTg AD mice. Our analysis of the hippocampus and cortical subregions revealed microvascular degeneration with onset and progression that was subregion dependent.


Sujet(s)
Vieillissement/métabolisme , Vieillissement/anatomopathologie , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Encéphale/vascularisation , Microvaisseaux/anatomopathologie , Plaque amyloïde/métabolisme , Maladie d'Alzheimer/imagerie diagnostique , Maladie d'Alzheimer/psychologie , Animaux , Encéphale/imagerie diagnostique , Cognition , Modèles animaux de maladie humaine , Évolution de la maladie , Mâle , Souches mutantes de souris , Souris transgéniques , Microvaisseaux/imagerie diagnostique , Microtomographie aux rayons X
3.
Front Aging Neurosci ; 12: 92, 2020.
Article de Anglais | MEDLINE | ID: mdl-32317959

RÉSUMÉ

Despite the extensive use of hormonal methods as either contraception or menopausal hormone therapy (HT), there is very little known about the potential effects of these compounds on the cellular processes of the brain. Medroxyprogesterone Acetate (MPA) is a progestogen used globally in the hormonal contraceptive, Depo Provera, by women in their reproductive prime and is a major compound found in HT formulations used by menopausal women. MPA promotes changes in the circulating levels of matrix metalloproteinases (MMPs), such as MMP-9, in the endometrium, yet limited literature studying the effects of MPA on neurons and astroglia cells has been conducted. Additionally, the dysregulation of MMPs has been implicated in the pathology of Alzheimer's disease (AD), where inhibiting the secretion of MMP-9 from astroglia reduces the proteolytic degradation of amyloid-beta. Thus, we hypothesize that exposure to MPA disrupts proteolytic degradation of amyloid-beta through the downregulation of MMP-9 expression and subsequent secretion. To assess the effect of progestins on MMP-9 and amyloid-beta, in vitro, C6 rat glial cells were exposed to MPA for 48 h and then the enzymatic, secretory, and amyloid-beta degrading capacity of MMP-9 was assessed from the conditioned culture medium. We found that MPA treatment inhibited transcription of MMP-9, which resulted in a subsequent decrease in the production and secretion of MMP-9 protein, in part through the glucocorticoid receptor. Additionally, we investigated the consequences of amyloid beta-degrading activity and found that MPA treatment decreased proteolytic degradation of amyloid-beta. Our results suggest MPA suppresses amyloid-beta degradation in an MMP-9-dependent manner, in vitro, and potentially compromises the clearance of amyloid-beta in vivo.

4.
J Alzheimers Dis ; 75(1): 119-138, 2020.
Article de Anglais | MEDLINE | ID: mdl-32250296

RÉSUMÉ

Cerebrovascular pathology is pervasive in Alzheimer's disease (AD), yet it is unknown whether cerebrovascular dysfunction contributes to the progression or etiology of AD. In human subjects and in animal models of AD, cerebral hypoperfusion and hypometabolism are reported to manifest during the early stages of the disease and persist for its duration. Amyloid-ß is known to cause cellular injury in both neurons and endothelial cells by inducing the production of reactive oxygen species and disrupting intracellular Ca2+ homeostasis. We present a mechanism for mitochondrial degeneration caused by the production of mitochondrial superoxide, which is driven by increased mitochondrial Ca2+ uptake. We found that persistent superoxide production injures mitochondria and disrupts electron transport in cerebrovascular endothelial cells. These observations provide a mechanism for the mitochondrial deficits that contribute to cerebrovascular dysfunction in patients with AD.


Sujet(s)
Peptides bêta-amyloïdes/pharmacologie , Calcium/métabolisme , Cellules endothéliales/métabolisme , Mitochondries/métabolisme , Fragments peptidiques/pharmacologie , Superoxydes/métabolisme , Régulation positive/effets des médicaments et des substances chimiques , Animaux , Encéphale/effets des médicaments et des substances chimiques , Encéphale/métabolisme , Lignée cellulaire , Cellules endothéliales/effets des médicaments et des substances chimiques , Souris , Mitochondries/effets des médicaments et des substances chimiques , Phosphorylation oxydative , Espèces réactives de l'oxygène/métabolisme
5.
J Alzheimers Dis ; 70(1): 139-151, 2019.
Article de Anglais | MEDLINE | ID: mdl-31177221

RÉSUMÉ

Mitochondrial dysfunction is often found in Alzheimer's disease (AD) patients and animal models. Clinical severity of AD is linked to early deficiencies in cognitive function and brain metabolism, indicating that pathological changes may begin early in life. Previous studies showed decreased mitochondrial function in primary hippocampal neurons from triple-transgenic Alzheimer's disease (3xTg-AD) mice and mitochondrial movement and structure deficits in primary neurons exposed to amyloid-ß oligomers. The present study characterized mitochondrial movement, number, and structure in 3xTg-AD primary cortical neurons and non-transgenic (nonTg) controls. We found a significant reduction in mitochondrial number and movement in 3xTg-AD primary cortical neurons with modest structural changes. Additionally, application of the sigma-1 receptor agonist, (+)SKF-10,047, markedly increased mitochondrial movement in both 3xTg-AD and nonTg primary cortical cultures after one hour of treatment. (+)SKF-10,047 also led to a trend of increased mitochondrial number in 3xTg-AD cultures. Embryonic mitochondrial movement and number deficits could be among the key steps in the early pathogenesis of AD that compromise cognitive or metabolic reserve, and amelioration of these deficits could be a promising area for further preclinical and clinical study.


Sujet(s)
Maladie d'Alzheimer/métabolisme , Cortex cérébral/métabolisme , Mitochondries/métabolisme , Neurones/métabolisme , Maladie d'Alzheimer/anatomopathologie , Animaux , Cortex cérébral/anatomopathologie , Modèles animaux de maladie humaine , Souris , Souris transgéniques , Mitochondries/anatomopathologie , Dynamique mitochondriale/physiologie , Neurones/anatomopathologie
6.
Mitochondrion ; 47: 244-255, 2019 07.
Article de Anglais | MEDLINE | ID: mdl-30594729

RÉSUMÉ

Astrocytes serve to maintain proper neuronal function and support neuronal viability, but remain largely understudied in research of cerebral ischemia. Astrocytic mitochondria are core participants in the metabolic activity of astrocytes. The objective of this study is to assess astrocyte mitochondrial competence during hypoxia and post-hypoxia reoxygenation and to determine cellular adaptive and pathological changes in the mitochondrial network. We hypothesize that during metabolic distress in astrocytes; mitochondrial networks undergo a shift in fission-fusion dynamics that results in a change in the morphometric state of the entire mitochondrial network. This mitochondrial network shift may be protective during metabolic distress by priming mitochondrial size and facilitating mitophagy. We demonstrated that hypoxia and post-hypoxia reoxygenation of rat primary astrocytes results in a redistribution of mitochondria to smaller sizes evoked by increased mitochondrial fission. Excessive mitochondrial fission corresponded to Drp-1 dephosphorylation at Ser 637, which preceded mitophagy of relatively small mitochondria. Reoxygenation of astrocytes marked the initiation of elevated mitophagic activity primarily reserved to the perinuclear region where a large number of the smallest mitochondria occurred. Although, during hypoxia astrocytic ATP content was severely reduced, after reoxygenation ATP content returned to near normoxic values and these changes mirrored mitochondrial superoxide production. Concomitant with these changes in astrocytic mitochondria, the number of astrocytic extensions declined only after 10-hours post-hypoxic reoxygenation. Overall, we posit a drastic mitochondrial network change that is triggered by a metabolic crisis during hypoxia; these changes are followed by mitochondrial degradation and retraction of astrocytic extensions during reoxygenation.


Sujet(s)
Astrocytes/métabolisme , Mitochondries/métabolisme , Dynamique mitochondriale , Mitophagie , Oxygène/métabolisme , Animaux , Astrocytes/anatomopathologie , Hypoxie cellulaire , Cellules cultivées , Dynamines/métabolisme , Mitochondries/anatomopathologie , Protéines mitochondriales/métabolisme , Oxygène/pharmacologie , Rats
7.
Metab Brain Dis ; 33(6): 2039-2044, 2018 12.
Article de Anglais | MEDLINE | ID: mdl-30267298

RÉSUMÉ

Chronic cerebrovascular hypoperfusion results in vascular dementia and increases predisposition to lacunar infarcts. However, there are no suitable animal models. In this study, we developed a novel model for chronic irreversible cerebral hypoperfusion in mice. Briefly, an ameroid constrictor was placed on the right carotid artery to gradually occlude the vessel, while a microcoil was placed on the left carotid artery to prevent compensation of the blood flow. This procedure resulted in a gradual hypoperfusion developing over a period of 34 days with no cerebral blood flow recovery. Histological analysis of the brain revealed neuronal and axonal degeneration as well as necrotic lesions. The most severely affected regions were located in the hippocampus and the corpus callosum. Overall, our paradigm is a viable model to study brain pathology resulting from gradual cerebrovascular hypoperfusion.


Sujet(s)
Artère carotide commune/anatomopathologie , Sténose carotidienne/anatomopathologie , Circulation cérébrovasculaire/physiologie , Démence vasculaire/anatomopathologie , Modèles animaux de maladie humaine , Animaux , Artère carotide commune/physiopathologie , Sténose carotidienne/complications , Sténose carotidienne/physiopathologie , Démence vasculaire/étiologie , Démence vasculaire/physiopathologie , Mâle , Souris , Souris de lignée C57BL
8.
J Cereb Blood Flow Metab ; 36(2): 387-92, 2016 Feb.
Article de Anglais | MEDLINE | ID: mdl-26661155

RÉSUMÉ

The blood-brain barrier is composed of cerebrovascular endothelial cells and tight junctions, and maintaining its integrity is crucial for the homeostasis of the neuronal environment. Recently, we discovered that mitochondria play a critical role in maintaining blood-brain barrier integrity. We report for the first time a novel mechanism underlying blood-brain barrier integrity: miR-34a mediated regulation of blood-brain barrier through a mitochondrial mechanism. Bioinformatics analysis suggests miR-34a targets several mitochondria-associated gene candidates. We demonstrated that miR-34a triggers the breakdown of blood-brain barrier in cerebrovascular endothelial cell monolayer in vitro, paralleled by reduction of mitochondrial oxidative phosphorylation and adenosine triphosphate production, and decreased cytochrome c levels.


Sujet(s)
Barrière hémato-encéphalique/physiologie , Cytochromes c/génétique , Cytochromes c/physiologie , microARN/génétique , Mitochondries/génétique , Mitochondries/physiologie , ADP/biosynthèse , Animaux , Circulation cérébrovasculaire/génétique , Circulation cérébrovasculaire/physiologie , Biologie informatique , Cellules endothéliales , Endothélium vasculaire/cytologie , Cinétique , Souris , Phosphorylation oxydative , Consommation d'oxygène/physiologie , Perméabilité , Jonctions serrées
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE