Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 4 de 4
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Fluids Barriers CNS ; 21(1): 42, 2024 May 16.
Article de Anglais | MEDLINE | ID: mdl-38755642

RÉSUMÉ

BACKGROUND: Most subarachnoid hemorrhage (SAH) patients have no obvious hematoma lesions but exhibit blood-brain barrier dysfunction and vasogenic brain edema. However, there is a few days between blood‒brain barrier dysfunction and vasogenic brain edema. The present study sought to investigate whether this phenomenon is caused by endothelial injury induced by the acute astrocytic barrier, also known as the glial limitans. METHODS: Bioinformatics analyses of human endothelial cells and astrocytes under hypoxia were performed based on the GEO database. Wild-type, EGLN3 and PKM2 conditional knock-in mice were used to confirm glial limitan formation after SAH. Then, the effect of endothelial EGLN3-PKM2 signaling on temporal and spatial changes in glial limitans was evaluated in both in vivo and in vitro models of SAH. RESULTS: The data indicate that in the acute phase after SAH, astrocytes can form a temporary protective barrier, the glia limitans, around blood vessels that helps maintain barrier function and improve neurological prognosis. Molecular docking studies have shown that endothelial cells and astrocytes can promote glial limitans-based protection against early brain injury through EGLN3/PKM2 signaling and further activation of the PKC/ERK/MAPK signaling pathway in astrocytes after SAH. CONCLUSION: Improving the ability to maintain glial limitans may be a new therapeutic strategy for improving the prognosis of SAH patients.


Sujet(s)
Astrocytes , Barrière hémato-encéphalique , Cellules endothéliales , Hypoxia-inducible factor-proline dioxygenases , Pyruvate kinase , Transduction du signal , Hémorragie meningée , Animaux , Humains , Souris , Astrocytes/métabolisme , Barrière hémato-encéphalique/métabolisme , Oedème cérébral/métabolisme , Cellules endothéliales/métabolisme , Protéines membranaires/métabolisme , Souris de lignée C57BL , Souris transgéniques , Pyruvate kinase/métabolisme , Transduction du signal/physiologie , Hémorragie meningée/métabolisme , Hémorragie meningée/immunologie , Hypoxia-inducible factor-proline dioxygenases/métabolisme
2.
J Neuroinflammation ; 21(1): 102, 2024 Apr 18.
Article de Anglais | MEDLINE | ID: mdl-38637850

RÉSUMÉ

The notion that the central nervous system is an immunologically immune-exempt organ has changed over the past two decades, with increasing evidence of strong links and interactions between the central nervous system and the peripheral immune system, both in the healthy state and after ischemic and hemorrhagic stroke. Although primary injury after stroke is certainly important, the limited therapeutic efficacy, poor neurological prognosis and high mortality have led researchers to realize that secondary injury and damage may also play important roles in influencing long-term neurological prognosis and mortality and that the neuroinflammatory process in secondary injury is one of the most important influences on disease progression. Here, we summarize the interactions of the central nervous system with the peripheral immune system after ischemic and hemorrhagic stroke, in particular, how the central nervous system activates and recruits peripheral immune components, and we review recent advances in corresponding therapeutic approaches and clinical studies, emphasizing the importance of the role of the peripheral immune system in ischemic and hemorrhagic stroke.


Sujet(s)
Lésions encéphaliques , Encéphalopathie ischémique , Tumeurs du cerveau , Accident vasculaire cérébral hémorragique , Accident vasculaire cérébral , Humains , Accident vasculaire cérébral hémorragique/complications , Encéphalopathie ischémique/complications , Encéphale , Accident vasculaire cérébral/complications , Lésions encéphaliques/complications , Tumeurs du cerveau/complications
3.
Cell Mol Neurobiol ; 44(1): 33, 2024 Apr 16.
Article de Anglais | MEDLINE | ID: mdl-38625414

RÉSUMÉ

Subarachnoid hemorrhage (SAH) is associated with high mortality and disability rates, and secondary white matter injury is an important cause of poor prognosis. However, whether brain capillary pericytes can directly affect the differentiation and maturation of oligodendrocyte precursor cells (OPCs) and subsequently affect white matter injury repair has still been revealed. This study was designed to investigate the effect of tissue inhibitor of metalloproteinase-3 (TIMP-3) for OPC differentiation and maturation. PDGFRßret/ret and wild-type C57B6J male mice were used to construct a mouse model of SAH via endovascular perforation in this study. Mice were also treated with vehicle, TIMP-3 RNAi or TIMP-3 RNAi + TIMP-3 after SAH. The effect of TIMP-3 on the differentiation and maturation of OPCs was determined using behavioral score, ELISA, transmission electron microscopy, immunofluorescence staining and cell culture. We found that TIMP-3 was secreted mainly by pericytes and that SAH and TIMP-3 RNAi caused a significant decrease in the TIMP-3 content, reaching a nadir at 24 h, followed by gradual recovery. In vitro, the myelin basic protein content of oligodendrocytes after oxyhemoglobin treatment was increased by TIMP-3 overexpression. The data indicates TIMP-3 could promote the differentiation and maturation of OPCs and subsequently improve neurological outcomes after SAH. Therefore, TIMP-3 could be beneficial for repair after white matter injury and could be a potential therapeutic target in SAH.


Sujet(s)
Précurseurs des oligodendrocytes , Hémorragie meningée , Substance blanche , Mâle , Animaux , Souris , Inhibiteur tissulaire de métalloprotéinase-3 , Encéphale
4.
Exp Neurol ; 360: 114293, 2023 02.
Article de Anglais | MEDLINE | ID: mdl-36493862

RÉSUMÉ

BACKGROUND: Most patients with subarachnoid hemorrhage (SAH) do not exhibit brain parenchymal injury upon imaging but present significant blood-brain barrier (BBB) disruption and secondary neurological deficits. The aim of this study was to investigate whether stressed astrocytes act as a secondary barrier to exert a protective effect after SAH and to investigate the mechanism of glial limitan formation. METHODS: A total of 204 adult male C57BL/6 mice and an endovascular perforation SAH model were employed. The spatiotemporal characteristics of glial limitan formation after SAH were determined by immunofluorescence staining and transmission electron microscopy. The molecular mechanisms by which pericytes regulate glia limitans formation were analyzed using polymerase chain reaction, Western blotting, immunofluorescence staining and ELISA in a pericyte-astrocyte contact coculture system. The findings were validated ex vivo and in vivo using lentiviruses and inhibitors. Finally, pericytes were targeted to regulate glial limitan formation, and the effect of the glia limitans on secondary brain injury after SAH was evaluated by flow cytometry and analysis of neurological function. RESULTS: Stress-induced glial limitan formation occurred 1 day after SAH and markedly subsided 3 days after ictus. Pericytes regulated astrocyte glia limitan formation via EphA4/EphrinB2 signaling, inhibited inflammatory cell infiltration and altered neurological function. CONCLUSIONS: Astrocyte-derived glia limitans serve as a secondary protective barrier following BBB disruption after SAH in mice, and pericytes can regulate glial limitan formation and alter neurological function via EphA4/EphrinB2 signaling. Strategies for maintaining this secondary protective barrier may be novel treatment approaches for alleviating early brain injury after SAH.


Sujet(s)
Oedème cérébral , Lésions encéphaliques , Hémorragie meningée , Animaux , Mâle , Souris , Astrocytes , Barrière hémato-encéphalique , Éphrine B2 , Souris de lignée C57BL , Péricytes , Hémorragie meningée/complications , Récepteur EphA4/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE