Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.823
Filtrer
1.
Fluids Barriers CNS ; 21(1): 51, 2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38858667

RÉSUMÉ

Oedema occurs when higher than normal amounts of solutes and water accumulate in tissues. In brain parenchymal tissue, vasogenic oedema arises from changes in blood-brain barrier permeability, e.g. in peritumoral oedema. Cytotoxic oedema arises from excess accumulation of solutes within cells, e.g. ischaemic oedema following stroke. This type of oedema is initiated when blood flow in the affected core region falls sufficiently to deprive brain cells of the ATP needed to maintain ion gradients. As a consequence, there is: depolarization of neurons; neural uptake of Na+ and Cl- and loss of K+; neuronal swelling; astrocytic uptake of Na+, K+ and anions; swelling of astrocytes; and reduction in ISF volume by fluid uptake into neurons and astrocytes. There is increased parenchymal solute content due to metabolic osmolyte production and solute influx from CSF and blood. The greatly increased [K+]isf triggers spreading depolarizations into the surrounding penumbra increasing metabolic load leading to increased size of the ischaemic core. Water enters the parenchyma primarily from blood, some passing into astrocyte endfeet via AQP4. In the medium term, e.g. after three hours, NaCl permeability and swelling rate increase with partial opening of tight junctions between blood-brain barrier endothelial cells and opening of SUR1-TPRM4 channels. Swelling is then driven by a Donnan-like effect. Longer term, there is gross failure of the blood-brain barrier. Oedema resolution is slower than its formation. Fluids without colloid, e.g. infused mock CSF, can be reabsorbed across the blood-brain barrier by a Starling-like mechanism whereas infused serum with its colloids must be removed by even slower extravascular means. Large scale oedema can increase intracranial pressure (ICP) sufficiently to cause fatal brain herniation. The potentially lethal increase in ICP can be avoided by craniectomy or by aspiration of the osmotically active infarcted region. However, the only satisfactory treatment resulting in retention of function is restoration of blood flow, providing this can be achieved relatively quickly. One important objective of current research is to find treatments that increase the time during which reperfusion is successful. Questions still to be resolved are discussed.


Sujet(s)
Oedème cérébral , Encéphale , Humains , Oedème cérébral/physiopathologie , Oedème cérébral/métabolisme , Oedème cérébral/étiologie , Animaux , Encéphale/métabolisme , Encéphale/physiopathologie , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/physiopathologie , Encéphalopathie ischémique/physiopathologie , Encéphalopathie ischémique/métabolisme
2.
Int J Mol Sci ; 25(12)2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38928258

RÉSUMÉ

Aquaporins (AQPs), particularly AQP4, play a crucial role in regulating fluid dynamics in the brain, impacting the development and resolution of edema following traumatic brain injury (TBI). This review examines the alterations in AQP expression and localization post-injury, exploring their effects on brain edema and overall injury outcomes. We discuss the underlying molecular mechanisms regulating AQP expression, highlighting potential therapeutic strategies to modulate AQP function. These insights provide a comprehensive understanding of AQPs in TBI and suggest novel approaches for improving clinical outcomes through targeted interventions.


Sujet(s)
Aquaporines , Lésions traumatiques de l'encéphale , Lésions traumatiques de l'encéphale/métabolisme , Humains , Animaux , Aquaporines/métabolisme , Oedème cérébral/métabolisme , Oedème cérébral/étiologie , Aquaporine-4/métabolisme , Hydrodynamique , Encéphale/métabolisme
3.
Neuroreport ; 35(11): 679-686, 2024 Aug 07.
Article de Anglais | MEDLINE | ID: mdl-38874950

RÉSUMÉ

Intracerebral hemorrhage (ICH) is a significant public health matter that has no effective treatment. ICH-induced destruction of the blood-brain barrier (BBB) leads to neurological deterioration. Astrocytic sonic hedgehog (SHH) alleviates brain injury by maintaining the integrity of the BBB after ICH. Silent information regulator 1 (SIRT1) is neuroprotective in several central nervous system diseases via BBB regulation. It is also a possible influential factor of the SHH signaling pathway. Nevertheless, the role of SIRT1 on BBB and the underlying pathological process associated with the SHH signaling pathway after ICH remain unclear. We established an intracerebral hemorrhagic mouse model by collagenase injection. SRT1720 (a selective agonist of SIRT1) was used to evaluate the effect of SIRT1 on BBB integrity after ICH. SIRT1 expression was reduced in the mouse brain after ICH. SRT1720 attenuated neurobehavioral impairments and brain edema of ICH mouse. After ICH induction, SRT1720 improved BBB integrity and tight junction expressions in the mouse brain. The SHH signaling pathway-related factors smoothened and glioma-associated oncogene homolog-1 were increased with the intervention of SRT1720, while cyclopamine (a specific inhibitor of the SHH signaling pathway) reversed these effects. These findings suggest that SIRT1 protects from ICH by altering BBB permeability and tight junction expression levels. This process is associated with the SHH signaling pathway, suggesting that SIRT1 may be a potential therapeutic target for ICH.


Sujet(s)
Barrière hémato-encéphalique , Hémorragie cérébrale , Composés hétérocycliques avec 4 noyaux ou plus , Sirtuine-1 , Animaux , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Barrière hémato-encéphalique/métabolisme , Sirtuine-1/métabolisme , Hémorragie cérébrale/métabolisme , Hémorragie cérébrale/traitement médicamenteux , Composés hétérocycliques avec 4 noyaux ou plus/pharmacologie , Mâle , Souris , Modèles animaux de maladie humaine , Souris de lignée C57BL , Neuroprotecteurs/pharmacologie , Protéines Hedgehog/métabolisme , Protéines Hedgehog/agonistes , Oedème cérébral/traitement médicamenteux , Oedème cérébral/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
4.
J Physiol ; 602(13): 3151-3168, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38924526

RÉSUMÉ

Aquaporin-4 (AQP4) is the main water channel in brain and is enriched in perivascular astrocyte processes abutting brain microvessels. There is a rich literature on the role of AQP4 in experimental stroke. While its role in oedema formation following middle cerebral artery occlusion (MCAO) has been studied extensively, its specific impact on infarct volume remains unclear. This study investigated the effects of total and partial AQP4 deletion on infarct volume in mice subjected to distal medial cerebral artery (dMCAO) occlusion. Compared to MCAO, this model induces smaller infarcts confined to neocortex, and less oedema. We show that AQP4 deletion significantly reduced infarct volume as assessed 1 week after dMCAO, suggesting that the role of AQP4 in stroke goes beyond its effect on oedema formation and dissolution. The reduction in infarct volume was associated with increased astrocyte reactivity in the peri-infarct areas. No significant differences were observed in the number of microglia among the genotypes. These findings provide new insights in the role of AQP4 in ischaemic injury indicating that AQP4 affects both infarct volume and astrocyte reactivity in the peri-infarct zone. KEY POINTS: Aquaporin-4 (AQP4) is the main water channel in brain and is enriched in perivascular astrocyte processes abutting microvessels. A rich literature exists on the role of AQP4 in oedema formation following middle cerebral artery occlusion (MCAO). We investigated the effects of total and partial AQP4 deletion on infarct volume in mice subjected to distal medial cerebral artery occlusion (dMCAO), a model inducing smaller infarcts confined to neocortex and less oedema compared to MCAO. AQP4 deletion significantly reduced infarct volume 1 week after dMCAO, suggesting a broader role for AQP4 in stroke beyond oedema formation. The reduction in infarct volume was associated with increased astrocyte reactivity in the peri-infarct areas, while no significant differences were observed in the number of microglia among the genotypes. These findings provide new insights into the role of AQP4 in stroke, indicating that AQP4 affects both infarct volume and astrocyte reactivity in the peri-infarct zone.


Sujet(s)
Aquaporine-4 , Astrocytes , Animaux , Aquaporine-4/génétique , Aquaporine-4/métabolisme , Astrocytes/métabolisme , Astrocytes/anatomopathologie , Souris , Mâle , Infarctus du territoire de l'artère cérébrale moyenne/métabolisme , Infarctus du territoire de l'artère cérébrale moyenne/génétique , Infarctus du territoire de l'artère cérébrale moyenne/anatomopathologie , Infarctus du territoire de l'artère cérébrale moyenne/physiopathologie , Souris de lignée C57BL , Modèles animaux de maladie humaine , Accident vasculaire cérébral/anatomopathologie , Accident vasculaire cérébral/métabolisme , Accident vasculaire cérébral/génétique , Souris knockout , Oedème cérébral/anatomopathologie , Oedème cérébral/métabolisme , Oedème cérébral/génétique
5.
BMC Cardiovasc Disord ; 24(1): 266, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38773462

RÉSUMÉ

BACKGROUND: Cardiopulmonary bypass (CPB) results in brain injury, which is primarily caused by inflammation. Ac2-26 protects against ischemic or hemorrhage brain injury. The present study was to explore the effect and mechanism of Ac2-26 on brain injury in CPB rats. METHODS: Forty-eight rats were randomized into sham, CPB, Ac, Ac/AKT1, Ac/GSK3ßi and Ac/AKT1/GSK3ßa groups. Rats in sham group only received anesthesia and in the other groups received standard CPB surgery. Rats in the sham and CPB groups received saline, and rats in the Ac, Ac/AKT1, Ac/GSK3ßi and Ac/AKT1/GSK3ßa groups received Ac2-26 immediately after CPB. Rats in the Ac/AKT1, Ac/GSK3ßi and Ac/AKT1/GSK3ßa groups were injected with shRNA, inhibitor and agonist of GSK3ß respectively. The neurological function score, brain edema and histological score were evaluated. The neuronal survival and hippocampal pyroptosis were assessed. The cytokines, activity of NF-κB, S100 calcium-binding protein ß(S100ß) and neuron-specific enolase (NSE), and oxidative were tested. The NLRP3, cleaved-caspase-1 and cleaved-gadermin D (GSDMD) in the brain were also detected. RESULTS: Compared to the sham group, all indicators were aggravated in rats that underwent CPB. Compared to the CPB group, Ac2-26 significantly improved neurological scores and brain edema and ameliorated pathological injury. Ac2-26 reduced the local and systemic inflammation, oxidative stress response and promoted neuronal survival. Ac2-26 reduced hippocampal pyroptosis and decreased pyroptotic proteins in brain tissue. The protection of Ac2-26 was notably lessened by shRNA and inhibitor of GSK3ß. The agonist of GSK3ß recovered the protection of Ac2-26 in presence of shRNA. CONCLUSIONS: Ac2-26 significantly improved neurological function, reduced brain injury via regulating inflammation, oxidative stress response and pyroptosis after CPB. The protective effect of Ac2-26 primarily depended on AKT1/ GSK3ß pathway.


Sujet(s)
Pontage cardiopulmonaire , Modèles animaux de maladie humaine , Glycogen synthase kinase 3 beta , Protéines proto-oncogènes c-akt , Pyroptose , Rat Sprague-Dawley , Transduction du signal , Animaux , Pontage cardiopulmonaire/effets indésirables , Glycogen synthase kinase 3 beta/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Pyroptose/effets des médicaments et des substances chimiques , Mâle , Neurones/effets des médicaments et des substances chimiques , Neurones/anatomopathologie , Neurones/métabolisme , Neurones/enzymologie , Neuroprotecteurs/pharmacologie , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Oedème cérébral/prévention et contrôle , Oedème cérébral/métabolisme , Oedème cérébral/enzymologie , Oedème cérébral/anatomopathologie , Anti-inflammatoires/pharmacologie , Rats , Sous-unité bêta de la protéine liant le calcium S100/métabolisme , Médiateurs de l'inflammation/métabolisme
6.
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 , Transduction du signal , Hémorragie meningée , Animaux , Astrocytes/métabolisme , Humains , Hémorragie meningée/métabolisme , Hémorragie meningée/immunologie , Souris , Transduction du signal/physiologie , Barrière hémato-encéphalique/métabolisme , Cellules endothéliales/métabolisme , Souris de lignée C57BL , Mâle , Pyruvate kinase/métabolisme , Protéines de transport/métabolisme , Oedème cérébral/métabolisme , Souris transgéniques , Protéines membranaires/métabolisme
7.
Sci Rep ; 14(1): 11585, 2024 05 21.
Article de Anglais | MEDLINE | ID: mdl-38773195

RÉSUMÉ

High-altitude cerebral edema (HACE) is a severe neurological condition that can occur at high altitudes. It is characterized by the accumulation of fluid in the brain, leading to a range of symptoms, including severe headache, confusion, loss of coordination, and even coma and death. Exosomes play a crucial role in intercellular communication, and their contents have been found to change in various diseases. This study analyzed the metabolomic characteristics of blood exosomes from HACE patients compared to those from healthy controls (HCs) with the aim of identifying specific metabolites or metabolic pathways associated with the development of HACE conditions. A total of 21 HACE patients and 21 healthy controls were recruited for this study. Comprehensive metabolomic profiling of the serum exosome samples was conducted using ultraperformance liquid chromatography-tandem mass spectrometry (UPLC‒MS/MS). Additionally, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis was performed to identify the metabolic pathways affected in HACE patients. Twenty-six metabolites, including ( +)-camphoric acid, choline, adenosine, adenosine 5'-monophosphate, deoxyguanosine 5'-monophosphate, guanosine, and hypoxanthine-9-ß-D-arabinofuranoside, among others, exhibited significant changes in expression in HACE patients compared to HCs. Additionally, these differentially abundant metabolites were confirmed to be potential biomarkers for HACE. KEGG pathway enrichment analysis revealed several pathways that significantly affect energy metabolism regulation (such as purine metabolism, thermogenesis, and nucleotide metabolism), estrogen-related pathways (the estrogen signaling pathway, GnRH signaling pathway, and GnRH pathway), cyclic nucleotide signaling pathways (the cGMP-PKG signaling pathway and cAMP signaling pathway), and hormone synthesis and secretion pathways (renin secretion, parathyroid hormone synthesis, secretion and action, and aldosterone synthesis and secretion). In patients with HACE, adenosine, guanosine, and hypoxanthine-9-ß-D-arabinofuranoside were negatively correlated with height. Deoxyguanosine 5'-monophosphate is negatively correlated with weight and BMI. Additionally, LPE (18:2/0:0) and pregnanetriol were positively correlated with age. This study identified potential biomarkers for HACE and provided valuable insights into the underlying metabolic mechanisms of this disease. These findings may lead to potential targets for early diagnosis and therapeutic intervention in HACE patients.


Sujet(s)
Marqueurs biologiques , Oedème cérébral , Exosomes , Métabolomique , Humains , Mâle , Femelle , Adulte , Métabolomique/méthodes , Oedème cérébral/sang , Oedème cérébral/métabolisme , Oedème cérébral/étiologie , Marqueurs biologiques/sang , Exosomes/métabolisme , Spectrométrie de masse en tandem , Mal de l'altitude/sang , Mal de l'altitude/métabolisme , Adulte d'âge moyen , Voies et réseaux métaboliques , Métabolome , Études cas-témoins , Altitude
8.
J Neuroinflammation ; 21(1): 140, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38807233

RÉSUMÉ

BACKGROUND: Perihematomal edema (PHE) after post-intracerebral hemorrhage (ICH) has complex pathophysiological mechanisms that are poorly understood. The complicated immune response in the post-ICH brain constitutes a crucial component of PHE pathophysiology. In this study, we aimed to characterize the transcriptional profiles of immune cell populations in human PHE tissue and explore the microscopic differences between different types of immune cells. METHODS: 9 patients with basal ganglia intracerebral hemorrhage (hematoma volume 50-100 ml) were enrolled in this study. A multi-stage profile was developed, comprising Group1 (n = 3, 0-6 h post-ICH, G1), Group2 (n = 3, 6-24 h post-ICH, G2), and Group3 (n = 3, 24-48 h post-ICH, G3). A minimal quantity of edematous tissue surrounding the hematoma was preserved during hematoma evacuation. Single cell RNA sequencing (scRNA-seq) was used to map immune cell populations within comprehensively resected PHE samples collected from patients at different stages after ICH. RESULTS: We established, for the first time, a comprehensive landscape of diverse immune cell populations in human PHE tissue at a single-cell level. Our study identified 12 microglia subsets and 5 neutrophil subsets in human PHE tissue. What's more, we discovered that the secreted phosphoprotein-1 (SPP1) pathway served as the basis for self-communication between microglia subclusters during the progression of PHE. Additionally, we traced the trajectory branches of different neutrophil subtypes. Finally, we also demonstrated that microglia-produced osteopontin (OPN) could regulate the immune environment in PHE tissue by interacting with CD44-positive cells. CONCLUSIONS: As a result of our research, we have gained valuable insight into the immune-microenvironment within PHE tissue, which could potentially be used to develop novel treatment modalities for ICH.


Sujet(s)
Oedème cérébral , Hémorragie cérébrale , Évolution de la maladie , Analyse de séquence d'ARN , Analyse sur cellule unique , Humains , Oedème cérébral/immunologie , Oedème cérébral/anatomopathologie , Oedème cérébral/génétique , Oedème cérébral/métabolisme , Oedème cérébral/étiologie , Hémorragie cérébrale/immunologie , Hémorragie cérébrale/anatomopathologie , Hémorragie cérébrale/génétique , Mâle , Femelle , Adulte d'âge moyen , Analyse de séquence d'ARN/méthodes , Sujet âgé , Hématome/anatomopathologie , Hématome/immunologie , Hématome/génétique
9.
J Stroke Cerebrovasc Dis ; 33(7): 107738, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38701940

RÉSUMÉ

OBJECTIVES: Edaravone dexborneol is neuroprotective against ischemic stroke, with free radical-scavenging and anti-inflammatory effects, but its effects in hemorrhagic stroke remain unclear. We evaluated whether edaravone dexborneol has a neuroprotective effect in intracerebral hemorrhage, and its underlying mechanisms. MATERIALS AND METHODS: Bioinformatics were used to predict the pathway of action of edaravone dexborneol. An intracerebral hemorrhage model was established using type IV collagenase in edaravone dexborneol, intracerebral hemorrhage, Sham, adeno-associated virus + edaravone dexborneol, and adeno-associated virus + intracerebral hemorrhage groups. The modified Neurological Severity Score was used to evaluate neurological function in rats. Brain water content was measured using the dry-wet weight method. Tumor necrosis factor-α, interleukin-1ß, inducible nitric oxide synthase, and γ-aminobutyric acid levels were determined by enzyme-linked immunosorbent assay. The expression levels of neurofilament light chain and γ-aminobutyric acid transaminase were determined by western blot. Nissl staining was used to examine neuronal morphology. Cognitive behavior was evaluated using a small-animal treadmill. RESULTS: Edaravone dexborneol alleviated neurological defects, improved cognitive function, and reduced cerebral edema, neuronal degeneration, and necrosis in rats with cerebral hemorrhage. The expression levels of neurofilament light chain, tumor necrosis factor-α, interleukin-1ß, inducible nitric oxide synthase, and γ-aminobutyric acid were decreased, while γ-aminobutyric acid transaminase expression was up-regulated. CONCLUSIONS: Edaravone dexborneol regulates γ-aminobutyric acid content by acting on the γ-aminobutyric acid transaminase signaling pathway, thus alleviating oxidative stress, neuroinflammation, neuronal degeneration, and death caused by excitatory toxic injury of neurons after intracerebral hemorrhage.


Sujet(s)
Oedème cérébral , Modèles animaux de maladie humaine , Édaravone , Interleukine-1 bêta , Neuroprotecteurs , Rat Sprague-Dawley , Animaux , Édaravone/pharmacologie , Mâle , Neuroprotecteurs/pharmacologie , Interleukine-1 bêta/métabolisme , Oedème cérébral/anatomopathologie , Oedème cérébral/traitement médicamenteux , Oedème cérébral/métabolisme , Oedème cérébral/enzymologie , Oedème cérébral/prévention et contrôle , 4-Aminobutyrate transaminase/métabolisme , 4-Aminobutyrate transaminase/antagonistes et inhibiteurs , Comportement animal/effets des médicaments et des substances chimiques , Hémorragie cérébrale/traitement médicamenteux , Hémorragie cérébrale/métabolisme , Hémorragie cérébrale/anatomopathologie , Hémorragie cérébrale/enzymologie , Anti-inflammatoires/pharmacologie , Cognition/effets des médicaments et des substances chimiques , Encéphale/effets des médicaments et des substances chimiques , Encéphale/anatomopathologie , Encéphale/métabolisme , Encéphale/enzymologie , Nitric oxide synthase type II/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Facteur de nécrose tumorale alpha/métabolisme , Médiateurs de l'inflammation/métabolisme
10.
Resuscitation ; 200: 110243, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38796092

RÉSUMÉ

BACKGROUND: Selective water uptake by neurons and glial cells and subsequent brain tissue oedema are key pathophysiological processes of hypoxic-ischemic encephalopathy (HIE) after cardiac arrest (CA). Although brain computed tomography (CT) is widely used to assess the severity of HIE, changes of brain radiodensity over time have not been investigated. These could be used to quantify regional brain net water uptake (NWU), a potential prognostic biomarker. METHODS: We conducted an observational prognostic accuracy study including a derivation (single center cardiac arrest registry) and a validation (international multicenter TTM2 trial) cohort. Early (<6 h) and follow-up (>24 h) head CTs of CA patients were used to determine regional NWU for grey and white matter regions after co-registration with a brain atlas. Neurological outcome was dichotomized as good versus poor using the Cerebral Performance Category Scale (CPC) in the derivation cohort and Modified Rankin Scale (mRS) in the validation cohort. RESULTS: We included 115 patients (81 derivation, 34 validation) with out-of-hospital (OHCA) and in-hospital cardiac arrest (IHCA). Regional brain water content remained unchanged in patients with good outcome. In patients with poor neurological outcome, we found considerable regional water uptake with the strongest effect in the basal ganglia. NWU >8% in the putamen and caudate nucleus predicted poor outcome with 100% specificity (95%-CI: 86-100%) and 43% (moderate) sensitivity (95%-CI: 31-56%). CONCLUSION: This pilot study indicates that NWU derived from serial head CTs is a promising novel biomarker for outcome prediction after CA. NWU >8% in basal ganglia grey matter regions predicted poor outcome while absence of NWU indicated good outcome. NWU and follow-up CTs should be investigated in larger, prospective trials with standardized CT acquisition protocols.


Sujet(s)
Marqueurs biologiques , Tomodensitométrie , Humains , Mâle , Femelle , Adulte d'âge moyen , Tomodensitométrie/méthodes , Sujet âgé , Pronostic , Marqueurs biologiques/métabolisme , Marqueurs biologiques/analyse , Arrêt cardiaque hors hôpital/thérapie , Arrêt cardiaque hors hôpital/imagerie diagnostique , Arrêt cardiaque/métabolisme , Encéphale/imagerie diagnostique , Encéphale/métabolisme , Hypoxie-ischémie du cerveau/imagerie diagnostique , Hypoxie-ischémie du cerveau/métabolisme , Oedème cérébral/étiologie , Oedème cérébral/imagerie diagnostique , Oedème cérébral/métabolisme , Enregistrements
11.
Biomed Pharmacother ; 176: 116834, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38815288

RÉSUMÉ

Although diabetes mellitus negatively affects post-ischaemic stroke injury and recovery, its impact on intracerebral haemorrhage (ICH) remains uncertain. This study aimed to investigate the effect of experimental diabetes (ED) on ICH-induced injury and neurological impairment. Sprague-Dawley rats were induced with ED 2 weeks before ICH induction. Animals were randomly assigned to four groups: 1)Healthy; 2)ICH; 3)ED; 4)ED-ICH. ICH and ED-ICH groups showed similar functional assessment. The ED-ICH group exhibited significantly lower haemorrhage volume compared with the ICH group, except at 1 mo. The oedema/ICH volume ratio and cistern displacement ratio were significantly higher in the ED-ICH group. Vascular markers revealed greater expression of α-SMA in the ED groups (ED and ED-ICH) compared with ICH. Conversely, the ICH groups (ED-ICH and ICH) exhibited higher levels of VEGF compared to the healthy and ED groups. An assessment of myelin tract integrity showed an increase in fractional anisotropy in the ED and ED-ICH groups compared with ICH. The ED group showed higher cryomyelin expression than the ED-ICH and ICH groups. Additionally, the ED groups (ED and ED-ICH) displayed higher expression of MOG and Olig-2 than ICH. As for inflammation, MCP-1 levels were significantly lower in the ED-ICH groups compared with the ICH group. Notably, ED did not aggravate the neurological outcome; however, it results in greater ICH-related brain oedema, greater brain structure displacement and lower haemorrhage volume. ED influences the cerebral vascularisation with an increase in vascular thickness, limits the inflammatory response and attenuates the deleterious effect of ICH on white matter integrity.


Sujet(s)
Hémorragie cérébrale , Diabète expérimental , Rat Sprague-Dawley , Animaux , Hémorragie cérébrale/anatomopathologie , Hémorragie cérébrale/métabolisme , Mâle , Diabète expérimental/complications , Diabète expérimental/métabolisme , Rats , Oedème cérébral/anatomopathologie , Oedème cérébral/métabolisme , Oedème cérébral/étiologie , Modèles animaux de maladie humaine , Encéphale/métabolisme , Encéphale/anatomopathologie
12.
J Neuroinflammation ; 21(1): 106, 2024 Apr 24.
Article de Anglais | MEDLINE | ID: mdl-38658922

RÉSUMÉ

BACKGROUND: Intracerebral hemorrhage (ICH) is a devastating neurological disease causing severe sensorimotor dysfunction and cognitive decline, yet there is no effective treatment strategy to alleviate outcomes of these patients. The Mas axis-mediated neuroprotection is involved in the pathology of various neurological diseases, however, the role of the Mas receptor in the setting of ICH remains to be elucidated. METHODS: C57BL/6 mice were used to establish the ICH model by injection of collagenase into mice striatum. The Mas receptor agonist AVE0991 was administered intranasally (0.9 mg/kg) after ICH. Using a combination of behavioral tests, Western blots, immunofluorescence staining, hematoma volume, brain edema, quantitative-PCR, TUNEL staining, Fluoro-Jade C staining, Nissl staining, and pharmacological methods, we examined the impact of intranasal application of AVE0991 on hematoma absorption and neurological outcomes following ICH and investigated the underlying mechanism. RESULTS: Mas receptor was found to be significantly expressed in activated microglia/macrophages, and the peak expression of Mas receptor in microglia/macrophages was observed at approximately 3-5 days, followed by a subsequent decline. Activation of Mas by AVE0991 post-treatment promoted hematoma absorption, reduced brain edema, and improved both short- and long-term neurological functions in ICH mice. Moreover, AVE0991 treatment effectively attenuated neuronal apoptosis, inhibited neutrophil infiltration, and reduced the release of inflammatory cytokines in perihematomal areas after ICH. Mechanistically, AVE0991 post-treatment significantly promoted the transformation of microglia/macrophages towards an anti-inflammatory, phagocytic, and reparative phenotype, and this functional phenotypic transition of microglia/macrophages by Mas activation was abolished by both Mas inhibitor A779 and Nrf2 inhibitor ML385. Furthermore, hematoma clearance and neuroprotective effects of AVE0991 treatment were reversed after microglia depletion in ICH. CONCLUSIONS: Mas activation can promote hematoma absorption, ameliorate neurological deficits, alleviate neuron apoptosis, reduced neuroinflammation, and regulate the function and phenotype of microglia/macrophages via Akt/Nrf2 signaling pathway after ICH. Thus, intranasal application of Mas agonist ACE0991 may provide promising strategy for clinical treatment of ICH patients.


Sujet(s)
Hématome , Accident vasculaire cérébral hémorragique , Souris de lignée C57BL , Récepteurs couplés aux protéines G , Récupération fonctionnelle , Animaux , Souris , Hématome/traitement médicamenteux , Hématome/anatomopathologie , Hématome/métabolisme , Mâle , Accident vasculaire cérébral hémorragique/anatomopathologie , Accident vasculaire cérébral hémorragique/traitement médicamenteux , Récepteurs couplés aux protéines G/agonistes , Récepteurs couplés aux protéines G/métabolisme , Récupération fonctionnelle/effets des médicaments et des substances chimiques , Récupération fonctionnelle/physiologie , Protéines proto-oncogènes/métabolisme , Oedème cérébral/étiologie , Oedème cérébral/métabolisme , Oedème cérébral/traitement médicamenteux , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme
13.
Brain Res ; 1834: 148907, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38570153

RÉSUMÉ

BACKGROUND: Traumatic brain injury (TBI), as a major public health problem, is characterized by high incidence rate, disability rate, and mortality rate. Neuroinflammation plays a crucial role in the pathogenesis of TBI. Triggering receptor expressed on myeloid cells-1 (TREM-1) is recognized as an amplifier of the inflammation in diseases of the central nervous system (CNS). However, the function of TREM-1 remains unclear post-TBI. This study aimed to investigate the function of TREM-1 in neuroinflammation induced by TBI. METHODS: Brain water content (BWC), modified neurological severity score (mNSS), and Morris Water Maze (MWM) were measured to evaluate the effect of TREM-1 inhibition on nervous system function and outcome after TBI. TREM-1 expression in vivo was evaluated by Western blotting. The cellular localization of TREM-1 in the damaged region was observed via immunofluorescence staining. We also conducted Western blotting to examine expression of SYK, p-SYK and other downstream proteins. RESULTS: We found that inhibition of TREM-1 reduced brain edema, decreased mNSS and improved neurobehavioral outcomes after TBI. It was further determined that TREM-1 was expressed on microglia and modulated subtype transition of microglia. Inhibition of TREM-1 alleviated neuroinflammation, which was associated with SYK/p38MAPK signaling pathway. CONCLUSIONS: These findings suggest that TREM-1 can be a potential clinical therapeutic target for alleviating neuroinflammation after TBI.


Sujet(s)
Lésions traumatiques de l'encéphale , Microglie , Maladies neuro-inflammatoires , Syk kinase , Récepteur de déclenchement de type-1 exprimé sur les cellules myéloïdes , p38 Mitogen-Activated Protein Kinases , Lésions traumatiques de l'encéphale/métabolisme , Lésions traumatiques de l'encéphale/traitement médicamenteux , Animaux , Récepteur de déclenchement de type-1 exprimé sur les cellules myéloïdes/métabolisme , Récepteur de déclenchement de type-1 exprimé sur les cellules myéloïdes/antagonistes et inhibiteurs , Microglie/métabolisme , Microglie/effets des médicaments et des substances chimiques , Syk kinase/métabolisme , Syk kinase/antagonistes et inhibiteurs , Mâle , Maladies neuro-inflammatoires/métabolisme , Maladies neuro-inflammatoires/traitement médicamenteux , p38 Mitogen-Activated Protein Kinases/métabolisme , Souris , Transduction du signal/effets des médicaments et des substances chimiques , Oedème cérébral/métabolisme , Oedème cérébral/traitement médicamenteux , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Système de signalisation des MAP kinases/physiologie , Souris de lignée C57BL
14.
Aging (Albany NY) ; 16(8): 6990-7008, 2024 04 10.
Article de Anglais | MEDLINE | ID: mdl-38613810

RÉSUMÉ

BACKGROUND: Intracerebral hemorrhage (ICH) comprises primary and secondary injuries, the latter of which induces increased inflammation and apoptosis and is more severe. Activating transcription factor 6 (ATF6) is a type-II transmembrane protein in the endoplasmic reticulum (ER). ATF6 target genes could improve ER homeostasis, which contributes to cryoprotection. Hence, we predict that ATF6 will have a protective effect on brain tissue after ICH. METHOD: The ICH rat model was generated through autologous blood injection into the right basal ganglia, the expression of ATF6 after ICH was determined by WB and IF. The expression of ATF6 was effectively controlled by means of intervention, and a series of measures was used to detect cell death, neuroinflammation, brain edema, blood-brain barrier and other indicators after ICH. Finally, the effects on long-term neural function of rats were measured by behavioral means. RESULT: ATF6 was significantly increased in the ICH-induced brain tissues. Further, ATF6 was found to modulate the expression of cystathionine γ-lyase (CTH) after ICH. Upregulation of ATF6 attenuated neuronal apoptosis and inflammation in ICH rats, along with mitigation of ICH-induced brain edema, blood-brain barrier deterioration, and cognitive behavior defects. Conversely, ATF6 genetic knockdown induced effects counter to those aforementioned. CONCLUSIONS: This study thereby emphasizes the crucial role of ATF6 in secondary brain injury in response to ICH, indicating that ATF6 upregulation may potentially ameliorate ICH-induced secondary brain injury. Consequently, ATF6 could serve as a promising therapeutic target to alleviate clinical ICH-induced secondary brain injuries.


Sujet(s)
Facteur de transcription ATF-6 , Hémorragie cérébrale , Cystathionine gamma-lyase , Animaux , Mâle , Rats , Facteur de transcription ATF-6/métabolisme , Facteur de transcription ATF-6/génétique , Apoptose , Barrière hémato-encéphalique/métabolisme , Encéphale/métabolisme , Encéphale/anatomopathologie , Oedème cérébral/métabolisme , Lésions encéphaliques/métabolisme , Hémorragie cérébrale/métabolisme , Cystathionine gamma-lyase/métabolisme , Cystathionine gamma-lyase/génétique , Modèles animaux de maladie humaine , Rat Sprague-Dawley
15.
Int Immunopharmacol ; 131: 111869, 2024 Apr 20.
Article de Anglais | MEDLINE | ID: mdl-38492343

RÉSUMÉ

BACKGROUND AND PURPOSE: It has been reported activation of NLRP3 inflammasome after intracerebral hemorrhage (ICH) ictus exacerbates neuroinflammation and brain injury. We hypothesized that inhibition of NLRP3 by OLT1177 (dapansutrile), a novel NLRP3 inflammasome inhibitor, could reduce brain edema and attenuate brain injury in experimental ICH. METHODS: ICH was induced by injection of autologous blood into basal ganglia in mice models. Sixty-three C57Bl/6 male mice were randomly grouped into the sham, vehicle, OLT1177 (Dapansutrile, 200 mg/kg intraperitoneally) and treated for consecutive three days, starting from 1 h after ICH surgery. Behavioral test, brain edema, brain water content, blood-brain barrier integrity and vascular permeability, cell apoptosis, and NLRP3 and its downstream protein levels were measured. RESULTS: OLT1177 significantly reduced cerebral edema after ICH and contributed to the attenuation of neurological deficits. OLT1177 could preserve blood-brain barrier integrity and lessen vascular leakage. In addition, OLT1177 preserved microglia morphological shift and significantly inhibited the activation of caspase-1 and release of IL-1ß. We also found that OLT1177 can protect against neuronal loss in the affected hemisphere. CONCLUSIONS: OLT1177 (dapansutrile) could significantly attenuate the brain edema after ICH and effectively alleviate the neurological deficit. This result suggests that the novel NLRP3 inhibitor, OLT1177, might serve as a promising candidate for the treatment of ICH.


Sujet(s)
Oedème cérébral , Lésions encéphaliques , Nitriles , Sulfones , Souris , Mâle , Animaux , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Inflammasomes/métabolisme , Oedème cérébral/traitement médicamenteux , Oedème cérébral/métabolisme , Hémorragie cérébrale/traitement médicamenteux , Hémorragie cérébrale/métabolisme , Lésions encéphaliques/métabolisme
16.
Neuropharmacology ; 251: 109896, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38490299

RÉSUMÉ

Secondary brain injury after intracerebral hemorrhage (ICH) is the main cause of poor prognosis in ICH patients, but the underlying mechanisms remain less known. The involvement of Piezo1 in brain injury after ICH was studied in a mouse model of ICH. ICH was established by injecting autologous arterial blood into the basal ganglia in mice. After vehicle, Piezo1 blocker, GsMTx4, Piezo1 activator, Yoda-1, or together with mannitol (tail vein injection) was injected into the left lateral ventricle of mouse brain, Piezo1 level and the roles of Piezo1 in neuronal injury, brain edema, and neurological dysfunctions after ICH were determined by the various indicated methods. Piezo1 protein level in neurons was significantly upregulated 24 h after ICH in vivo (human and mice). Piezo1 protein level was also dramatically upregulated in HT22 cells (a murine neuron cell line) cultured in vitro 24 h after hemin treatment as an in vitro ICH model. GsMTx4 treatment or together with mannitol significantly downregulated Piezo1 and AQP4 levels, markedly increased Bcl2 level, maintained more neurons alive, considerably restored brain blood flow, remarkably relieved brain edema, substantially decreased serum IL-6 level, and almost fully reversed the neurological dysfunctions at ICH 24 h group mice. In contrast, Yoda-1 treatment achieved the opposite effects. In conclusion, Piezo1 plays a crucial role in the pathogenesis of brain injury after ICH and may be a target for clinical treatment of ICH.


Sujet(s)
Oedème cérébral , Lésions encéphaliques , Pyrazines , Thiadiazoles , Humains , Souris , Animaux , Hémorragie cérébrale/complications , Lésions encéphaliques/traitement médicamenteux , Canaux ioniques , Oedème cérébral/métabolisme , Mannitol/usage thérapeutique
17.
Brain Res ; 1837: 148855, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38471644

RÉSUMÉ

Subarachnoid hemorrhage (SAH) is characterized by the extravasation of blood into the subarachnoid space, in which erythrocyte lysis is the primary contributor to cell death and brain injuries. New evidence has indicated that meningeal lymphatic vessels (mLVs) are essential in guiding fluid and macromolecular waste from cerebrospinal fluid (CSF) into deep cervical lymph nodes (dCLNs). However, the role of mLVs in clearing erythrocytes after SAH has not been completely elucidated. Hence, we conducted a cross-species study. Autologous blood was injected into the subarachnoid space of rabbits and rats to induce SAH. Erythrocytes in the CSF were measured with/without deep cervical lymph vessels (dCLVs) ligation. Additionally, prior to inducing SAH, we administered rats with vascular endothelial growth factor C (VEGF-C), which is essential for meningeal lymphangiogenesis and maintaining integrity and survival of lymphatic vessels. The results showed that the blood clearance rate was significantly lower after dCLVs ligation in both the rat and rabbit models. DCLVs ligation aggravated neuroinflammation, neuronal damage, brain edema, and behavioral impairment after SAH. Conversely, the treatment of VEGF-C enhanced meningeal lymphatic drainage of erythrocytes and improved outcomes in SAH. In summary, our research highlights the indispensable role of the meningeal lymphatic pathway in the clearance of blood and mediating consequences after SAH.


Sujet(s)
Vaisseaux lymphatiques , Rat Sprague-Dawley , Hémorragie meningée , Animaux , Lapins , Hémorragie meningée/métabolisme , Rats , Mâle , Ligature/méthodes , Érythrocytes/métabolisme , Modèles animaux de maladie humaine , Facteur de croissance endothéliale vasculaire de type C/métabolisme , Méninges , Oedème cérébral/métabolisme
18.
J Neurosurg ; 141(1): 100-107, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38335517

RÉSUMÉ

OBJECTIVE: CD44 is a major cell surface receptor involved in cell adhesion and migration. The overexpression of CD44 is a poor prognostic factor in many neoplasms, including meningiomas. The aim of this study was to investigate the association between CD44 gene expression and clinical signatures of primary meningiomas. METHODS: CD44 gene expression was quantitatively evaluated by snap freezing tumor tissues obtained from 106 patients with primary meningioma. The relationships between CD44 expression and clinical signatures of meningiomas, including histological malignancy, tumor volume, and peritumoral brain edema (PTBE), were analyzed. PTBE was assessed using the Steinhoff classification (SC) system (from SC 0 to SC III). RESULTS: CD44 gene expression in WHO grade 2 and 3 meningiomas was significantly higher than that in grade 1 meningiomas. In addition, CD44 expression increased with the severity of PTBE. Particularly, among the grade 1 meningiomas or small-sized tumors (maximum tumor diameter < 43 mm), CD44 expression in tumors with severe PTBE (SC II or III) was significantly higher than that in tumors without or with mild PTBE (SC 0 or I). Multivariate logistic regression analysis also revealed that overexpression of CD44 was an independent significant factor of severe PTBE development in primary meningiomas. CONCLUSIONS: In addition to tumor cell aggressiveness, CD44 expression promotes the development of PTBE in meningioma. Since PTBE is a strong factor of tumor-related epilepsy or cognitive dysfunction in patients with meningioma, CD44 is thus a potential therapeutic target in meningioma with PTBE.


Sujet(s)
Oedème cérébral , Antigènes CD44 , Tumeurs des méninges , Méningiome , Humains , Méningiome/métabolisme , Méningiome/complications , Méningiome/anatomopathologie , Méningiome/génétique , Antigènes CD44/métabolisme , Antigènes CD44/génétique , Oedème cérébral/métabolisme , Oedème cérébral/étiologie , Oedème cérébral/anatomopathologie , Mâle , Tumeurs des méninges/métabolisme , Tumeurs des méninges/anatomopathologie , Tumeurs des méninges/complications , Tumeurs des méninges/génétique , Femelle , Adulte d'âge moyen , Sujet âgé , Adulte , Sujet âgé de 80 ans ou plus , Pertinence clinique
19.
Mol Neurobiol ; 61(8): 5614-5627, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38217667

RÉSUMÉ

This study aimed to investigate the effects of G1-activated G protein-coupled estrogen receptor 1 (GPER1) on neurological impairments and neuroinflammation in traumatic brain injury (TBI) mice. The controlled cortical impingement (CCI) method was used to establish the TBI model. The mice were subjected to ovariectomy (OVX) for two weeks prior to modeling. GPER1 agonist G1 was administered by intracerebroventricular injection. Brain tissue water content was detected by wet/dry method, and blood-brain barrier damage was detected by Evans blue extravasation. The neurological impairments in mice were evaluated by open field test, Y-maze test, nest-building test, object location memory test and novel object recognition test. Ionized calcium-binding adapter molecule 1 (Iba1) staining was used to indicate the activation of microglia. Expression of M1/M2-type microglia markers and inflammatory factors were evaluated by ELISA and qRT-PCR. The G1 administration significantly reduced cerebral edema and Evans blue extravasation at injury ipsilateral cortex and basal ganglia in TBI mice. Activation of GPER1 by G1 improved the anxiety behavior and the cognitive dysfunction of mice induced by TBI. G1 administration significantly decreased Iba1-positive staining cells and the mRNA levels of CD86, macrophage cationic peptide 1 (Mcp-1), nitric oxide synthase 2 (Nos2), interleukin 1 beta (IL-1ß), and macrophage inflammatory protein-2 (MIP-2), while increased the mRNA levels of interleukin 10 (IL-10), arginase1 (Arg-1) and CD206. Activation of GPER1 through G1 administration has the potential to ameliorate cognitive dysfunction induced by TBI in mice. It may also inhibit the activation of M1 microglia in cortical tissue resulting from TBI, while promoting the activation of M2 microglia and contributing to the regulation of inflammatory responses.


Sujet(s)
Lésions traumatiques de l'encéphale , Microglie , Récepteurs couplés aux protéines G , Animaux , Lésions traumatiques de l'encéphale/complications , Lésions traumatiques de l'encéphale/métabolisme , Lésions traumatiques de l'encéphale/anatomopathologie , Récepteurs couplés aux protéines G/métabolisme , Souris , Femelle , Microglie/métabolisme , Microglie/effets des médicaments et des substances chimiques , Récepteurs des oestrogènes/métabolisme , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/anatomopathologie , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Maladies neuro-inflammatoires/étiologie , Maladies neuro-inflammatoires/métabolisme , Maladies neuro-inflammatoires/traitement médicamenteux , Souris de lignée C57BL , Maladies du système nerveux/étiologie , Oedème cérébral/métabolisme , Oedème cérébral/étiologie
20.
Metab Brain Dis ; 39(3): 403-437, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-37606786

RÉSUMÉ

Brain edema is considered as a common feature associated with hepatic encephalopathy (HE). However, its central role as cause or consequence of HE and its implication in the development of the neurological alterations linked to HE are still under debate. It is now well accepted that type A and type C HE are biologically and clinically different, leading to different manifestations of brain edema. As a result, the findings on brain edema/swelling in type C HE are variable and sometimes controversial. In the light of the changing natural history of liver disease, better description of the clinical trajectory of cirrhosis and understanding of molecular mechanisms of HE, and the role of brain edema as a central component in the pathogenesis of HE is revisited in the current review. Furthermore, this review highlights the main techniques to measure brain edema and their advantages/disadvantages together with an in-depth description of the main ex-vivo/in-vivo findings using cell cultures, animal models and humans with HE. These findings are instrumental in elucidating the role of brain edema in HE and also in designing new multimodal studies by performing in-vivo combined with ex-vivo experiments for a better characterization of brain edema longitudinally and of its role in HE, especially in type C HE where water content changes are small.


Sujet(s)
Oedème cérébral , Encéphalopathie hépatique , Animaux , Humains , Encéphalopathie hépatique/métabolisme , Oedème cérébral/métabolisme , Encéphale/métabolisme , Modèles animaux , Cirrhose du foie/complications
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...