Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Mol Neurobiol ; 43(2): 525-541, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35195811

RESUMO

Microvessels in the central nervous system (CNS) have one of the highest populations of pericytes, indicating their crucial role in maintaining homeostasis. Pericytes are heterogeneous cells located around brain microvessels; they present three different morphologies along the CNS vascular tree: ensheathing, mesh, and thin-strand pericytes. At the arteriole-capillary transition ensheathing pericytes are found, while mesh and thin-strand pericytes are located at capillary beds. Brain pericytes are essential for the establishment and maintenance of the blood-brain barrier, which restricts the passage of soluble and potentially toxic molecules from the circulatory system to the brain parenchyma. Pericytes play a key role in regulating local inflammation at the CNS. Pericytes can respond differentially, depending on the degree of inflammation, by secreting a set of neurotrophic factors to promote cell survival and regeneration, or by potentiating inflammation through the release of inflammatory mediators (e.g., cytokines and chemokines), and the overexpression of cell adhesion molecules. Under inflammatory conditions, pericytes may regulate immune cell trafficking to the CNS and play a role in perpetuating local inflammation. In this review, we describe pericyte responses during acute and chronic neuroinflammation.


Assuntos
Doenças Neuroinflamatórias , Pericitos , Adulto , Humanos , Encéfalo/irrigação sanguínea , Barreira Hematoencefálica , Sistema Nervoso Central
2.
Methods Mol Biol ; 2367: 37-46, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32789776

RESUMO

The study of the regionalized function of the blood-brain barrier at the level of brain endothelial cells and pericytes is essential to understand the biological properties and molecular mechanisms regulating this biological barrier. The isolation of blood vessels from specific brain regions will allow to understand regional differences in susceptibility to pathological phenomena such as ischemia, traumatic brain injury, and neurodegenerative diseases, such as Alzheimer disease. Here, we propose an efficient and fast method to isolate brain endothelial cells and pericytes from a specific cerebral region. The isolated brain endothelial cells and pericytes are viable to perform conventional molecular and histological techniques such as Western blots, immunocytofluorescence, and scanning electron microscopy.


Assuntos
Encéfalo , Barreira Hematoencefálica , Células Endoteliais , Microvasos , Pericitos
3.
Brain Behav Immun ; 89: 118-132, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32485292

RESUMO

Sleep loss in the rat increases blood-brain barrier permeability to circulating molecules by disrupting interendothelial tight junctions. Despite the description of the ultrastructure of cerebral microvessels and the evidence of an apparent pericyte detachment from capillary wall in sleep restricted rats the effect of sleep loss on pericytes is unknown. Here we characterized the interactions between pericytes and brain endothelial cells after sleep loss using male Wistar rats. Animals were sleep-restricted 20 h daily with 4 h sleep recovery for 10 days. At the end of the sleep restriction, brain microvessels (MVs) were isolated from cerebral cortex and hippocampus and processed for Western blot and immunocytochemistry to evaluate markers of pericyte-endothelial cell interaction (connexin 43, PDGFR-ß), tight junction proteins, and proinflammatory mediator proteins (MMP9, A2A adenosine receptor, CD73, NFκB). Sleep restriction reduced PDGFR-ß and connexin 43 expression in MVs; in addition, scanning electron microscopy micrographs showed that pericytes were detached from capillary walls, but did not undergo apoptosis (as depicted by a reduced active caspase-3 expression). Sleep restriction also decreased tight junction protein expression in MVs and increased BBB permeability to low- and high-molecular weight tracers in in vivo permeability assays. Those alterations seemed to depend on a low-grade inflammatory status as reflected by the increased expression of phosphorylated NFκB and A2A adenosine receptor in brain endothelial cells from the sleep-restricted rats. Our data show that pericyte-brain endothelial cell interaction is altered by sleep restriction; this evidence is essential to understand the role of sleep in regulating blood-brain barrier function.


Assuntos
Barreira Hematoencefálica , Pericitos , Animais , Encéfalo , Comunicação Celular , Células Endoteliais , Masculino , Ratos , Ratos Wistar , Sono , Junções Íntimas
4.
J Sleep Res ; 29(3): e12907, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31578801

RESUMO

Sleep loss increases blood-brain barrier permeability. As the blood-brain barrier and the blood-tissue barriers in the reproductive tract (blood-testis and blood-epididymis barriers) share common characteristics, we hypothesized that sleep restriction may also modify their barrier function. Previous reports showed that sleep loss decreased sperm viability and progressive fast mobility, which may be a consequence of altered blood-testis and blood-epididymis barrier. Therefore, we quantified changes in blood-testis and blood-epididymis barrier after sleep loss and related them to male fertility. Adult male Wistar rats were sleep restricted using the multiple-platform technique in a protocol of 20 hr daily sleep deprivation plus 4 hr of sleep recovery in the home-cage. At the 10th day, barrier permeability assays were performed with Na-fluorescein, 10 kDa Cascade blue-dextrans and Evans blue, and the expression of tight junction proteins, actin and androgen receptor was quantified. At the 10th day of sleep restriction and after sleep recovery days 1-7, males were placed with sexually receptive females, sexual behaviour was tested, and the percentage of pregnancies was calculated. Sleep restriction increased the barrier permeability to low- and high-molecular-weight tracers, and decreased the expression of tight junction proteins, actin and androgen receptor. Concomitantly, sleep restriction reduced the percentage of ejaculating males and the number of pregnancies. Sleep recovery for 2-3 days progressively re-established fertility, as indicated by a higher percentage of ejaculating males and impregnated females. In conclusion, chronic sleep loss alters fertility concomitantly with the disruption of the blood-tissue barriers at the reproductive tract, the mechanism involves androgen signalling.


Assuntos
Barreira Hematoencefálica/fisiopatologia , Epididimo/fisiopatologia , Fertilidade/fisiologia , Microscopia Confocal/métodos , Distúrbios do Início e da Manutenção do Sono/complicações , Animais , Doença Crônica , Humanos , Masculino , Ratos , Ratos Wistar , Privação do Sono/fisiopatologia , Testículo/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...