Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Fluids Barriers CNS ; 21(1): 42, 2024 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-38755642

RESUMO

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.


Assuntos
Astrócitos , Barreira Hematoencefálica , Células Endoteliais , Transdução de Sinais , Hemorragia Subaracnóidea , Animais , Astrócitos/metabolismo , Humanos , Hemorragia Subaracnóidea/metabolismo , Hemorragia Subaracnóidea/imunologia , Camundongos , Transdução de Sinais/fisiologia , Barreira Hematoencefálica/metabolismo , Células Endoteliais/metabolismo , Camundongos Endogâmicos C57BL , Masculino , Piruvato Quinase/metabolismo , Proteínas de Transporte/metabolismo , Edema Encefálico/metabolismo , Camundongos Transgênicos , Proteínas de Membrana/metabolismo
2.
Cell Mol Neurobiol ; 44(1): 33, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38625414

RESUMO

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.


Assuntos
Células Precursoras de Oligodendrócitos , Hemorragia Subaracnóidea , Substância Branca , Masculino , Animais , Camundongos , Inibidor Tecidual de Metaloproteinase-3 , Encéfalo
3.
J Transl Med ; 21(1): 196, 2023 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-36918921

RESUMO

BACKGROUND: Owing to metabolic disequilibrium and immune suppression, intracerebral hemorrhage (ICH) patients are prone to infections; according to a recent global analysis of stroke cases, approximately 10 million new-onset ICH patients had experienced concurrent infection. However, the intrinsic mechanisms underlying the effects of infection related peripheral inflammation after ICH remain unclear. METHODS: Lipopolysaccharide (LPS) was intraperitoneally injected into ICH model mice to induce peripheral inflammation. Neurobehavioral deficits, blood‒brain barrier (BBB) disruption, and the expression of CCR5, JAK2, STAT3, and MMP9 were evaluated after treatment with recombinant CCL5 (rCCL5) (a CCR5 ligand), maraviroc (MVC) (an FDA-approved selective CCR5 antagonist), or JAK2 CRISPR plasmids. RESULTS: Our study revealed that severe peripheral inflammation increased CCL5/CCR5 axis activation in multiple inflammatory cell types, including microglia, astrocytes, and monocytes, and aggravated BBB disruption and neurobehavioral dysfunction after ICH, possibly in part through the JAK2/STAT3 signaling pathway. CONCLUSIONS: CCR5 might be a potential target for the clinical treatment of infection-induced exacerbation of BBB disruption following ICH.


Assuntos
Barreira Hematoencefálica , Acidente Vascular Cerebral , Animais , Camundongos , Astrócitos , Hemorragia Cerebral/complicações , Hemorragia Cerebral/tratamento farmacológico , Hemorragia Cerebral/metabolismo , Inflamação/metabolismo , Acidente Vascular Cerebral/metabolismo
4.
Exp Neurol ; 360: 114293, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36493862

RESUMO

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.


Assuntos
Edema Encefálico , Lesões Encefálicas , Hemorragia Subaracnóidea , Animais , Masculino , Camundongos , Astrócitos , Barreira Hematoencefálica , Efrina-B2 , Camundongos Endogâmicos C57BL , Pericitos , Hemorragia Subaracnóidea/complicações , Receptor EphA4/metabolismo
5.
Transl Stroke Res ; 14(4): 443-445, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-35689126

RESUMO

Neutrophil extracellular traps (NETs) play a major role in intrinsic immunity by limiting and killing pathogens. Recently, a series of studies have confirmed that NETs are closely associated with vascular injury and microthrombosis. Furthermore, NETs play an important role in neuroinflammation after ischemic and hemorrhagic stroke. Neuroinflammation and microthrombosis after subarachnoid hemorrhage are key pathophysiological processes associated with poor prognosis, but their crucial formation mechanisms and interventions remain to be elucidated. Could NETs, as an emerging and important pathogenesis, be a new therapeutic target after subarachnoid hemorrhage?


Assuntos
Armadilhas Extracelulares , Hemorragia Subaracnóidea , Trombose , Humanos , Armadilhas Extracelulares/fisiologia , Hemorragia Subaracnóidea/complicações , Doenças Neuroinflamatórias , Trombose/tratamento farmacológico , Trombose/etiologia , Neutrófilos
6.
J Neurochem ; 160(1): 13-33, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34160835

RESUMO

Aneurysmal subarachnoid hemorrhage (aSAH) is a devastating cerebrovascular disease with high mortality and morbidity. In recent years, a large number of studies have focused on the mechanism of early brain injury (EBI) and delayed cerebral ischemia (DCI), including vasospasm, neurotoxicity of hematoma and neuroinflammatory storm, after aSAH. Despite considerable efforts, no novel drugs have significantly improved the prognosis of patients in phase III clinical trials, indicating the need to further re-examine the multifactorial pathophysiological process that occurs after aSAH. The complex pathogenesis is reflected by the destruction of the dynamic balance of the energy metabolism in the nervous system after aSAH, which prevents the maintenance of normal neural function. This review focuses on the fluid metabolic pathways of the central nervous system (CNS), starting with ruptured aneurysms, and discusses the dysfunction of blood circulation, cerebrospinal fluid (CSF) circulation and the glymphatic system during disease progression. It also proposes a hypothesis on the metabolic disorder mechanism and potential therapeutic targets for aSAH patients. Cover Image for this issue: https://doi.org/10.1111/jnc.15384.


Assuntos
Circulação Cerebrovascular/fisiologia , Sistema Glinfático/fisiologia , Redes e Vias Metabólicas/fisiologia , Hemorragia Subaracnóidea/metabolismo , Hemorragia Subaracnóidea/fisiopatologia , Animais , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Humanos
7.
Front Neurol ; 12: 659740, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34335439

RESUMO

Aneurysmal subarachnoid hemorrhage (SAH) is one of the special stroke subtypes with high mortality and mobility. Although the mortality of SAH has decreased by 50% over the past two decades due to advances in neurosurgery and management of neurocritical care, more than 70% of survivors suffer from varying degrees of neurological deficits and cognitive impairments, leaving a heavy burden on individuals, families, and the society. Recent studies have shown that white matter is vulnerable to SAH, and white matter injuries may be one of the causes of long-term neurological deficits caused by SAH. Attention has recently focused on the pivotal role of white matter injury in the pathophysiological processes after SAH, mainly related to mechanical damage caused by increased intracerebral pressure and the metabolic damage induced by blood degradation and hypoxia. In the present review, we sought to summarize the pathophysiology processes and mechanisms of white matter injury after SAH, with a view to providing new strategies for the prevention and treatment of long-term cognitive dysfunction after SAH.

8.
Exp Neurol ; 341: 113688, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33713655

RESUMO

Increasing numbers of patients with spontaneous subarachnoid hemorrhage(SAH) who recover from surgery and intensive care management still live with cognitive impairment after discharge, indicating the importance of white matter injury at the acute stage of SAH. In the present study, standard endovascular perforation was employed to establish an SAH mouse model, and a microRNA (miRNA) chip was used to analyze the changes in gene expression in white matter tissue after SAH. The data indicate that 17 miRNAs were downregulated, including miR-706, miR-669a-5p, miR-669p-5p, miR-7116-5p and miR-195a-3p, while 13 miRNAs were upregulated, including miR-6907-5p, miR-5135, miR-6982-5p, miR-668-5p, miR-8119. Strikingly, miR-706 was significantly downregulated with the highest fold change. Further experiments confirmed that miR-706 could alleviate white matter injury and improve neurological behavior, at least partially by inhibiting the PKCα/MST1/NF-κB pathway and the release of inflammatory cytokines. These results might provide a deeper understanding of the pathophysiological processes in white matter after SAH, as well as potential therapeutic strategies for the translational research.


Assuntos
Fator de Crescimento de Hepatócito/antagonistas & inibidores , MicroRNAs/biossíntese , NF-kappa B/antagonistas & inibidores , Proteína Quinase C-alfa/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Hemorragia Subaracnóidea/metabolismo , Substância Branca/metabolismo , Animais , Regulação para Baixo/fisiologia , Fator de Crescimento de Hepatócito/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/biossíntese , Proteína Quinase C-alfa/biossíntese , Proteínas Proto-Oncogênicas/biossíntese , Transdução de Sinais/fisiologia , Hemorragia Subaracnóidea/patologia , Hemorragia Subaracnóidea/prevenção & controle , Substância Branca/lesões , Substância Branca/patologia
9.
Transl Res ; 211: 139-146, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31103467

RESUMO

The minimally invasive surgery plus fibrinolysis has been identified as a promising treatment for spontaneous intracerebral hemorrhage (ICH). However, the fibrinolytic efficacy is not satisfactory. Neutrophil extracellular traps (NETs) have been demonstrated to impair fibrinolysis in sepsis and acute ischemic stroke. Therefore, we decided to explore the presence and potential effect of NETs in ICH fibrinolysis. Intracerebral hemorrhage was induced by autologous arterial blood injection into the basal ganglia in rats. First, at 0.5 hour, 1 hour, and 1.5 hours after blood injection, the brains were collected for NETs detection by immune-staining. Second, ICH rats were given intrahematoma fibrinolysis: rats were randomized to receive the equal amount of saline, DNAse 1, tissue-plasminogen activator (tPA), and tPA + DNAse 1 at 1 hour after hematoma placement. On day 3, animals were sacrificed for terminal deoxynucleotidyl transferase-mediated dUTP Nick-end labeling staining following MRI and behavioral tests. Third, on day 3 after ICH, the hematoma within brain were collected for ex vivo fibrinolysis assay to further evaluate the effect of NETs in ICH fibrinolysis. Co-staining of DAPI, H3, and MPO confirmed the presence of NETs in ICH. Disintegration of NETs using DNAse 1 enhanced tPA-induced hematoma fibrinolysis, relieved brain swelling, reduced cell death, and improved the functional outcome in ICH rats. Therefore, we concluded that NETs impaired the efficacy of tPA for ICH fibrinolysis in rats. Targeting NETs may be a new alternative to improve the fibrinolytic therapy following ICH.


Assuntos
Hemorragia Cerebral/terapia , Armadilhas Extracelulares , Ativador de Plasminogênio Tecidual , Animais , Fibrinolíticos/farmacologia , Distribuição Aleatória , Ratos
10.
Brain Res ; 1715: 41-46, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-30703371

RESUMO

Arginine-vasopressin (AVP) is believed to be positively correlated with the brain edema formation, but the underlying mechanism is still unclear. In this study, we explored the role of the V2 receptors antagonist tolvaptan on brain edema following intracerebral hemorrhage (ICH) with a rat model. Animals were randomly given tolvaptan or vehicle through oral gavage at 12 h, 36 h, and 60 h after ICH surgery. Brain swelling (%), brain water content(BWC), neurological scores, Evans blue fluorescence and blood-brain barrier (BBB) tight junction proteins were measured to evaluate the effect of tolvaptan in ICH. We found that tolvaptan alleviated the brain swelling (%), decreased the BWC growth, and attenuated the neurological deficits after ICH (p < 0.05, vs vehicle). What's more, tolvaptan increased the expression of ZO-1 and occludin (p < 0.05, vs vehicle), which might be attributed to the down-regulated effects of tolvaptan on MMP-9. These results provided evidence supporting the use of tolvaptan for ICH-induced brain edema. Large animal experiments are required to further explore the efficacy and mechanism of tolvaptan in ICH treatment.


Assuntos
Edema Encefálico/tratamento farmacológico , Hemorragia Cerebral/tratamento farmacológico , Tolvaptan/uso terapêutico , Animais , Transporte Biológico , Barreira Hematoencefálica/efeitos dos fármacos , Encéfalo/metabolismo , Edema Encefálico/metabolismo , Hemorragia Cerebral/metabolismo , Modelos Animais de Doenças , Masculino , Ocludina/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Vasopressinas/metabolismo
11.
J Immunol ; 202(5): 1441-1452, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30692211

RESUMO

Akt isoforms play key roles in multiple cellular processes; however, the roles of Akt-1 and Akt-2 isoforms in the development of T cell-mediated autoimmunity are poorly defined. In this study, we showed that Akt1-/- mice develop ameliorated experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis, whereas Akt2-/- mice develop exacerbated EAE, compared with wild-type mice. At the cellular level, Akt-1 appears to inhibit proliferation of thymus-derived regulatory T cells (tTregs), which facilitates Ag-specific Th1/Th17 responses. In a sharp contrast to Akt-1, Akt-2 potentiates tTreg proliferation in vitro and in vivo and suppresses Ag-specific Th1/Th17 responses. Furthermore, treating mice with established EAE with a specific Akt-1 inhibitor suppressed disease progression. Our data demonstrate that Akt-1 and Akt-2 differentially regulate the susceptibility of mice to EAE by controlling tTreg proliferation. Our data also indicate that targeting Akt-1 is a potential therapeutic approach for multiple sclerosis in humans.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Proteínas Proto-Oncogênicas c-akt/imunologia , Linfócitos T Reguladores/imunologia , Timo/imunologia , Animais , Proliferação de Células , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas c-akt/deficiência , Células Th1/imunologia , Células Th17/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA