Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 16.036
Filter
1.
J Physiol ; 602(13): 3151-3168, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38924526

ABSTRACT

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.


Subject(s)
Aquaporin 4 , Astrocytes , Animals , Aquaporin 4/genetics , Aquaporin 4/metabolism , Astrocytes/metabolism , Astrocytes/pathology , Mice , Male , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Mice, Inbred C57BL , Disease Models, Animal , Stroke/pathology , Stroke/metabolism , Stroke/genetics , Mice, Knockout , Brain Edema/pathology , Brain Edema/metabolism , Brain Edema/genetics
2.
CNS Neurosci Ther ; 30(6): e14796, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38867395

ABSTRACT

AIMS: The extent of perihematomal edema following intracerebral hemorrhage (ICH) significantly impacts patient prognosis, and disruption of the blood-brain barrier (BBB) exacerbates perihematomal edema. However, the role of peripheral IL-10 in mitigating BBB disruption through pathways that link peripheral and central nervous system signals remains poorly understood. METHODS: Recombinant IL-10 was administered to ICH model mice via caudal vein injection, an IL-10-inhibiting adeno-associated virus and an IL-10 receptor knockout plasmid were delivered intraventricularly, and neurobehavioral deficits, perihematomal edema, BBB disruption, and the expression of JAK1 and STAT3 were evaluated. RESULTS: Our study demonstrated that the peripheral cytokine IL-10 mitigated BBB breakdown, perihematomal edema, and neurobehavioral deficits after ICH and that IL-10 deficiency reversed these effects, likely through the IL-10R/JAK1/STAT3 signaling pathway. CONCLUSIONS: Peripheral IL-10 has the potential to reduce BBB damage and perihematomal edema following ICH and improve patient prognosis.


Subject(s)
Brain Edema , Cerebral Hemorrhage , Interleukin-10 , Janus Kinase 1 , Receptors, Interleukin-10 , STAT3 Transcription Factor , Signal Transduction , Animals , STAT3 Transcription Factor/metabolism , Cerebral Hemorrhage/complications , Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/metabolism , Brain Edema/etiology , Brain Edema/drug therapy , Janus Kinase 1/metabolism , Janus Kinase 1/antagonists & inhibitors , Interleukin-10/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism
3.
Aging (Albany NY) ; 16(11): 9959-9971, 2024 06 07.
Article in English | MEDLINE | ID: mdl-38850525

ABSTRACT

This meta-analysis aimed to describe the efficacy of bumetanide in improving infarct volume, brain edema, and behavioral outcomes in animal models of cerebral ischemia. Embase, PubMed and Web of Science databases were searched from their inception to February 2024 (INPLASY:202430023). Data on the animal species, stroke model, drug dose, time of treatment, method of administration, study quality, and outcomes were extracted and pooled in a meta-analysis. The combined standardized mean difference (SMD) or mean difference (MD) estimates and 95% confidence intervals (CIs) were calculated using random- or fixed-effects models. Thirteen eligible studies involving >200 animals fulfilled the inclusion criteria and were included in this meta-analysis. Meta-analyses demonstrated that bumetanide treatment significantly reduced cerebral infarct volume (SMD: -0.42; 95% CI: -0.75, -0.09; p < 0.01; n = 186 animals) and consistently relieved brain edema (SMD: -1.39; 95% CI: -2.06, -0.72; p < 0.01; n = 64 animals). Subgroup analyses demonstrated that bumetanide treatment reduced infarct volume in transient but not permanent cerebral ischemia models. When administered after the stroke, it was more effective than treatment initiation before the stroke. Eight studies assessed the effect of bumetanide on behavioral function and the results showed that bumetanide treatment significantly improved neurobehavioral deficits (SMD: -2.35; 95% CI: -2.72, -1.97; p < 0.01; n = 250 animals). We conclude that bumetanide appears to be effective in reducing infarct volume and brain edema and improving behavioral recovery in animal models of cerebral ischemia. This mechanism needs to be confirmed through further investigation.


Subject(s)
Bumetanide , Disease Models, Animal , Ischemic Stroke , Bumetanide/therapeutic use , Bumetanide/pharmacology , Animals , Ischemic Stroke/drug therapy , Ischemic Stroke/pathology , Brain Edema/drug therapy , Sodium Potassium Chloride Symporter Inhibitors/therapeutic use , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology
4.
Fluids Barriers CNS ; 21(1): 51, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38858667

ABSTRACT

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.


Subject(s)
Brain Edema , Brain , Humans , Brain Edema/physiopathology , Brain Edema/metabolism , Brain Edema/etiology , Animals , Brain/metabolism , Brain/physiopathology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/physiopathology , Brain Ischemia/physiopathology , Brain Ischemia/metabolism
5.
Arkh Patol ; 86(3): 52-58, 2024.
Article in Russian | MEDLINE | ID: mdl-38881006

ABSTRACT

Mucormycosis is a disease caused by fungi of the Mucorales family, widespread in the environment, with pronounced angiotropism and the ability to angioinvasion, leading to thrombosis with surrounding necrosis. The main triggers for the development of mucormycosis are: immunodeficiency states, use of glucocorticosteroid drugs, decompensation of diabetes mellitus, concomitant diseases, age > 65 years. We present a clinical case of rhinocerebral mucormycosis in a 79-year-old patient against the background of uncontrolled type 2 diabetes mellitus with ketoacidosis, a condition after previous glucocorticosteroid therapy for COVID-19 (according to the severity of the disease). After suffering a new coronavirus infection caused by the SARS-CoV-2 virus, she was admitted to the hospital with complaints characteristic of mucormycosis. On the 5th day of hospital stay, the patient's condition worsened significantly, despite the correction of the therapy, and on the 12th day the patient died. According to the results of the autopsy, it was established that the rhinocerebral mucormycosis was complicated by thrombosis of the anterior and posterior left cerebral arteries with subsequent infarctions in the frontal lobe and parieto-occipital region of the brain left hemisphere, cerebral edema, which was the immediate cause of death.


Subject(s)
COVID-19 , Diabetes Mellitus, Type 2 , Mucormycosis , Humans , Mucormycosis/diagnosis , Mucormycosis/microbiology , Mucormycosis/complications , Mucormycosis/etiology , Aged , Diabetes Mellitus, Type 2/complications , Female , Fatal Outcome , COVID-19/complications , SARS-CoV-2 , Brain Edema/microbiology , Brain Edema/etiology , Brain Edema/complications
6.
Nat Rev Dis Primers ; 10(1): 43, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38902312

ABSTRACT

Millions of people visit high-altitude regions annually and more than 80 million live permanently above 2,500 m. Acute high-altitude exposure can trigger high-altitude illnesses (HAIs), including acute mountain sickness (AMS), high-altitude cerebral oedema (HACE) and high-altitude pulmonary oedema (HAPE). Chronic mountain sickness (CMS) can affect high-altitude resident populations worldwide. The prevalence of acute HAIs varies according to acclimatization status, rate of ascent and individual susceptibility. AMS, characterized by headache, nausea, dizziness and fatigue, is usually benign and self-limiting, and has been linked to hypoxia-induced cerebral blood volume increases, inflammation and related trigeminovascular system activation. Disruption of the blood-brain barrier leads to HACE, characterized by altered mental status and ataxia, and increased pulmonary capillary pressure, and related stress failure induces HAPE, characterized by dyspnoea, cough and exercise intolerance. Both conditions are progressive and life-threatening, requiring immediate medical intervention. Treatment includes supplemental oxygen and descent with appropriate pharmacological therapy. Preventive measures include slow ascent, pre-acclimatization and, in some instances, medications. CMS is characterized by excessive erythrocytosis and related clinical symptoms. In severe CMS, temporary or permanent relocation to low altitude is recommended. Future research should focus on more objective diagnostic tools to enable prompt treatment, improved identification of individual susceptibilities and effective acclimatization and prevention options.


Subject(s)
Altitude Sickness , Altitude , Humans , Altitude Sickness/physiopathology , Altitude Sickness/epidemiology , Altitude Sickness/complications , Acclimatization/physiology , Brain Edema/physiopathology , Brain Edema/etiology , Brain Edema/epidemiology , Pulmonary Edema/physiopathology , Pulmonary Edema/etiology , Pulmonary Edema/epidemiology , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/epidemiology , Hypertension, Pulmonary/etiology , Hypoxia/physiopathology , Hypoxia/complications , Hypoxia/etiology
7.
Nutrients ; 16(12)2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38931235

ABSTRACT

Stroke is the world's second-leading cause of death. Current treatments for cerebral edema following intracerebral hemorrhage (ICH) mainly involve hyperosmolar fluids, but this approach is often inadequate. Propolis, known for its various beneficial properties, especially antioxidant and anti-inflammatory properties, could potentially act as an adjunctive therapy and help alleviate stroke-associated injuries. The chemical composition of Geniotrigona thoracica propolis extract was analyzed by GC-MS after derivatization for its total phenolic and total flavonoid content. The total phenolic content and total flavonoid content of the propolis extract were 1037.31 ± 24.10 µg GAE/mL and 374.02 ± 3.36 µg QE/mL, respectively. By GC-MS analysis, its major constituents were found to be triterpenoids (22.4% of TIC). Minor compounds, such as phenolic lipids (6.7% of TIC, GC-MS) and diterpenic acids (2.3% of TIC, GC-MS), were also found. Ninety-six Sprague Dawley rats were divided into six groups; namely, the control group, the ICH group, and four ICH groups that received the following therapies: mannitol, propolis extract (daily oral propolis administration after the ICH induction), propolis-M (propolis and mannitol), and propolis-B+A (daily oral propolis administration 7 days prior to and 72 h after the ICH induction). Neurocognitive functions of the rats were analyzed using the rotarod challenge and Morris water maze. In addition, the expression of NF-κB, SUR1-TRPM4, MMP-9, and Aquaporin-4 was analyzed using immunohistochemical methods. A TUNEL assay was used to assess the percentage of apoptotic cells. Mannitol significantly improved cognitive-motor functions in the ICH group, evidenced by improved rotarod and Morris water maze completion times, and lowered SUR-1 and Aquaporin-4 levels. It also significantly decreased cerebral edema by day 3. Similarly, propolis treatments (propolis-A and propolis-B+A) showed comparable improvements in these tests and reduced edema. Moreover, combining propolis with mannitol (propolis-M) further enhanced these effects, particularly in reducing edema and the Virchow-Robin space. These findings highlight the potential of propolis from the Indonesian stingless bee, Geniotrigona thoracica, from the Central Tapanuli region as a neuroprotective, adjunctive therapy.


Subject(s)
Cerebral Hemorrhage , Disease Models, Animal , Neuroprotective Agents , Propolis , Rats, Sprague-Dawley , Animals , Propolis/pharmacology , Propolis/chemistry , Neuroprotective Agents/pharmacology , Cerebral Hemorrhage/drug therapy , Bees , Rats , Male , Flavonoids/pharmacology , Flavonoids/analysis , Antioxidants/pharmacology , Brain Edema/drug therapy , Gas Chromatography-Mass Spectrometry , Phenols/pharmacology , Phenols/analysis
8.
Int J Mol Sci ; 25(12)2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38928258

ABSTRACT

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.


Subject(s)
Aquaporins , Brain Injuries, Traumatic , Brain Injuries, Traumatic/metabolism , Humans , Animals , Aquaporins/metabolism , Brain Edema/metabolism , Brain Edema/etiology , Aquaporin 4/metabolism , Hydrodynamics , Brain/metabolism
9.
Int J Mol Sci ; 25(12)2024 Jun 16.
Article in English | MEDLINE | ID: mdl-38928322

ABSTRACT

Despite continuous medical advancements, traumatic brain injury (TBI) remains a leading cause of death and disability worldwide. Consequently, there is a pursuit for biomarkers that allow non-invasive monitoring of patients after cranial trauma, potentially improving clinical management and reducing complications and mortality. Aquaporins (AQPs), which are crucial for transmembrane water transport, may be significant in this context. This study included 48 patients, with 27 having acute (aSDH) and 21 having chronic subdural hematoma (cSDH). Blood plasma samples were collected from the participants at three intervals: the first sample before surgery, the second at 15 h, and the third at 30 h post-surgery. Plasma concentrations of AQP1, AQP2, AQP4, and AQP9 were determined using the sandwich ELISA technique. CT scans were performed on all patients pre- and post-surgery. Correlations between variables were examined using Spearman's nonparametric rank correlation coefficient. A strong correlation was found between aquaporin 2 levels and the volume of chronic subdural hematoma and midline shift. However, no significant link was found between aquaporin levels (AQP1, AQP2, AQP4, and AQP9) before and after surgery for acute subdural hematoma, nor for AQP1, AQP4, and AQP9 after surgery for chronic subdural hematoma. In the chronic SDH group, AQP2 plasma concentration negatively correlated with the midline shift measured before surgery (Spearman's ρ -0.54; p = 0.017) and positively with hematoma volume change between baseline and 30 h post-surgery (Spearman's ρ 0.627; p = 0.007). No statistically significant correlation was found between aquaporin plasma levels and hematoma volume for AQP1, AQP2, AQP4, and AQP9 in patients with acute SDH. There is a correlation between chronic subdural hematoma volume, measured radiologically, and serum AQP2 concentration, highlighting aquaporins' potential as clinical biomarkers.


Subject(s)
Aquaporin 2 , Biomarkers , Brain Edema , Humans , Male , Female , Biomarkers/blood , Middle Aged , Aged , Prognosis , Brain Edema/blood , Brain Edema/etiology , Brain Edema/diagnostic imaging , Aquaporin 2/blood , Aquaporin 2/metabolism , Adult , Craniocerebral Trauma/blood , Craniocerebral Trauma/complications , Hematoma, Subdural, Chronic/blood , Hematoma, Subdural, Chronic/surgery , Aquaporin 1/blood , Aquaporin 1/metabolism , Tomography, X-Ray Computed , Brain Injuries, Traumatic/blood , Brain Injuries, Traumatic/diagnosis , Aquaporins/blood , Aquaporins/metabolism
10.
Neuroreport ; 35(11): 679-686, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-38874950

ABSTRACT

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.


Subject(s)
Blood-Brain Barrier , Cerebral Hemorrhage , Heterocyclic Compounds, 4 or More Rings , Sirtuin 1 , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Sirtuin 1/metabolism , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/drug therapy , Heterocyclic Compounds, 4 or More Rings/pharmacology , Male , Mice , Disease Models, Animal , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Hedgehog Proteins/metabolism , Hedgehog Proteins/agonists , Brain Edema/drug therapy , Brain Edema/metabolism , Signal Transduction/drug effects
11.
Trends Parasitol ; 40(7): 544-545, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38797655

ABSTRACT

A complex series of studies by Oelschlegel et al. in a murine model of cerebral malaria establishes a temporal sequence of events linking decreased venous efflux to impaired perfusion, edema, and neuroinflammation. The relevance to human cerebral malaria is discussed, including the heterogeneity recognized in recent investigations of cerebrovascular hemodynamics.


Subject(s)
Brain Edema , Disease Models, Animal , Malaria, Cerebral , Malaria, Cerebral/physiopathology , Malaria, Cerebral/parasitology , Animals , Mice , Brain Edema/parasitology , Humans
12.
PLoS Comput Biol ; 20(5): e1012145, 2024 May.
Article in English | MEDLINE | ID: mdl-38805558

ABSTRACT

In ischaemic stroke, a large reduction in blood supply can lead to the breakdown of the blood-brain barrier and to cerebral oedema after reperfusion therapy. The resulting fluid accumulation in the brain may contribute to a significant rise in intracranial pressure (ICP) and tissue deformation. Changes in the level of ICP are essential for clinical decision-making and therapeutic strategies. However, the measurement of ICP is constrained by clinical techniques and obtaining the exact values of the ICP has proven challenging. In this study, we propose the first computational model for the simulation of cerebral oedema following acute ischaemic stroke for the investigation of ICP and midline shift (MLS) relationship. The model consists of three components for the simulation of healthy blood flow, occluded blood flow and oedema, respectively. The healthy and occluded blood flow components are utilized to obtain oedema core geometry and then imported into the oedema model for the simulation of oedema growth. The simulation results of the model are compared with clinical data from 97 traumatic brain injury patients for the validation of major model parameters. Midline shift has been widely used for the diagnosis, clinical decision-making, and prognosis of oedema patients. Therefore, we focus on quantifying the relationship between ICP and midline shift (MLS) and identify the factors that can affect the ICP-MLS relationship. Three major factors are investigated, including the brain geometry, blood-brain barrier damage severity and the types of oedema (including rare types of oedema). Meanwhile, the two major types (stress and tension/compression) of mechanical brain damage are also presented and the differences in the stress, tension, and compression between the intraparenchymal and periventricular regions are discussed. This work helps to predict ICP precisely and therefore provides improved clinical guidance for the treatment of brain oedema.


Subject(s)
Brain Edema , Computer Simulation , Intracranial Pressure , Ischemic Stroke , Brain Edema/physiopathology , Humans , Ischemic Stroke/physiopathology , Ischemic Stroke/complications , Intracranial Pressure/physiology , Computational Biology , Cerebral Ventricles/physiopathology , Cerebral Ventricles/diagnostic imaging , Blood-Brain Barrier/physiopathology , Male
13.
J Stroke Cerebrovasc Dis ; 33(7): 107738, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38701940

ABSTRACT

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.


Subject(s)
Brain Edema , Disease Models, Animal , Edaravone , Interleukin-1beta , Neuroprotective Agents , Rats, Sprague-Dawley , Animals , Edaravone/pharmacology , Male , Neuroprotective Agents/pharmacology , Interleukin-1beta/metabolism , Brain Edema/pathology , Brain Edema/drug therapy , Brain Edema/metabolism , Brain Edema/enzymology , Brain Edema/prevention & control , 4-Aminobutyrate Transaminase/metabolism , 4-Aminobutyrate Transaminase/antagonists & inhibitors , Behavior, Animal/drug effects , Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/pathology , Cerebral Hemorrhage/enzymology , Anti-Inflammatory Agents/pharmacology , Cognition/drug effects , Brain/drug effects , Brain/pathology , Brain/metabolism , Brain/enzymology , Nitric Oxide Synthase Type II/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , Inflammation Mediators/metabolism
14.
Resuscitation ; 200: 110243, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38796092

ABSTRACT

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.


Subject(s)
Biomarkers , Tomography, X-Ray Computed , Humans , Male , Female , Middle Aged , Tomography, X-Ray Computed/methods , Aged , Prognosis , Biomarkers/metabolism , Biomarkers/analysis , Out-of-Hospital Cardiac Arrest/therapy , Out-of-Hospital Cardiac Arrest/diagnostic imaging , Heart Arrest/metabolism , Brain/diagnostic imaging , Brain/metabolism , Hypoxia-Ischemia, Brain/diagnostic imaging , Hypoxia-Ischemia, Brain/metabolism , Brain Edema/etiology , Brain Edema/diagnostic imaging , Brain Edema/metabolism , Registries
15.
Biomed Pharmacother ; 176: 116834, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38815288

ABSTRACT

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.


Subject(s)
Cerebral Hemorrhage , Diabetes Mellitus, Experimental , Rats, Sprague-Dawley , Animals , Cerebral Hemorrhage/pathology , Cerebral Hemorrhage/metabolism , Male , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Rats , Brain Edema/pathology , Brain Edema/metabolism , Brain Edema/etiology , Disease Models, Animal , Brain/metabolism , Brain/pathology
16.
Fluids Barriers CNS ; 21(1): 42, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755642

ABSTRACT

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.


Subject(s)
Astrocytes , Blood-Brain Barrier , Endothelial Cells , Signal Transduction , Subarachnoid Hemorrhage , Animals , Astrocytes/metabolism , Humans , Subarachnoid Hemorrhage/metabolism , Subarachnoid Hemorrhage/immunology , Mice , Signal Transduction/physiology , Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Mice, Inbred C57BL , Male , Pyruvate Kinase/metabolism , Carrier Proteins/metabolism , Brain Edema/metabolism , Mice, Transgenic , Membrane Proteins/metabolism
17.
Neurosciences (Riyadh) ; 29(2): 90-95, 2024 May.
Article in English | MEDLINE | ID: mdl-38740406

ABSTRACT

OBJECTIVES: To investigate the factors that contribute to the development of cerebral edema after aneurysm clipping in individuals with aneurysmal subarachnoid hemorrhage (aSAH). METHODS: A total of 232 patients with aSAH caused by rupture and treated with aneurysm clipping were included in the retrospective analysis of clinical data. Postoperatively, the participants were categorized into two groups based on the presence or absence of cerebral edema: a complication group (n=33) and a non-complication group (n=199).A comparison was made between the overall data of the 2 groups. RESULTS: In the complication group, there were higher proportions of patients experiencing recurrent bleeding, aneurysm in the posterior circulation, Fisher grade III-IV, World Federation of Neurosurgical Societies (WFNS) grade II, Hunt-Hess grade III-IV, concomitant hypertension, duration from onset to operation ≥12 h, and concomitant hematoma compared to the non-complication group (p<0.05). Cerebral edema after aneurysm clipping was associated with several risk factors including repeated bleeding, aneurysm in the back of the brain, Fisher grade III-IV, WFNS grade II, Hunt-Hess grade III-IV, simultaneous high blood pressure and hematoma, and a duration of at least 12 hours from the start of symptoms to the surgical procedure (p<0.05). CONCLUSION: In patients with aSAH, the risk of cerebral edema after aneurysm clipping is increased by recurrent bleeding, aneurysm in the posterior circulation, Fisher grade III-IV, WFNS grade II, Hunt-Hess grade III-IV, concomitant hypertension and hematoma, and duration of ≥12 h from onset to operation.


Subject(s)
Brain Edema , Subarachnoid Hemorrhage , Humans , Subarachnoid Hemorrhage/surgery , Subarachnoid Hemorrhage/complications , Male , Female , Middle Aged , Brain Edema/etiology , Risk Factors , Retrospective Studies , Adult , Aged , Postoperative Complications/epidemiology , Postoperative Complications/etiology , Intracranial Aneurysm/surgery , Intracranial Aneurysm/complications , Neurosurgical Procedures/adverse effects , Surgical Instruments/adverse effects , Aneurysm, Ruptured/surgery , Aneurysm, Ruptured/complications
18.
Sci Rep ; 14(1): 10008, 2024 05 01.
Article in English | MEDLINE | ID: mdl-38693282

ABSTRACT

Historically, investigators have not differentiated between patients with and without hemorrhagic transformation (HT) in large core ischemic stroke at risk for life-threatening mass effect (LTME) from cerebral edema. Our objective was to determine whether LTME occurs faster in those with HT compared to those without. We conducted a two-center retrospective study of patients with ≥ 1/2 MCA territory infarct between 2006 and 2021. We tested the association of time-to-LTME and HT subtype (parenchymal, petechial) using Cox regression, controlling for age, mean arterial pressure, glucose, tissue plasminogen activator, mechanical thrombectomy, National Institute of Health Stroke Scale, antiplatelets, anticoagulation, temperature, and stroke side. Secondary and exploratory outcomes included mass effect-related death, all-cause death, disposition, and decompressive hemicraniectomy. Of 840 patients, 358 (42.6%) had no HT, 403 (48.0%) patients had petechial HT, and 79 (9.4%) patients had parenchymal HT. LTME occurred in 317 (37.7%) and 100 (11.9%) had mass effect-related deaths. Parenchymal (HR 8.24, 95% CI 5.46-12.42, p < 0.01) and petechial HT (HR 2.47, 95% CI 1.92-3.17, p < 0.01) were significantly associated with time-to-LTME and mass effect-related death. Understanding different risk factors and sequelae of mass effect with and without HT is critical for informed clinical decisions.


Subject(s)
Hospitalization , Infarction, Middle Cerebral Artery , Humans , Female , Male , Aged , Retrospective Studies , Middle Aged , Infarction, Middle Cerebral Artery/complications , Cerebral Hemorrhage/etiology , Cerebral Hemorrhage/mortality , Cerebral Hemorrhage/complications , Brain Edema/etiology , Risk Factors , Ischemic Stroke/mortality
19.
Acta Neurochir (Wien) ; 166(1): 234, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38805034

ABSTRACT

PURPOSE: Progressive cerebral edema with refractory intracranial hypertension (ICP) requiring decompressive hemicraniectomy (DHC) is a severe manifestation of early brain injury (EBI) after aneurysmal subarachnoid hemorrhage (aSAH). The purpose of the study was to investigate whether a more pronounced cerebrospinal fluid (CSF) drainage has an influence on cerebral perfusion pressure (CPP) and the extent of EBI after aSAH. METHODS: Patients with aSAH and indication for ICP-monitoring admitted to our center between 2012 and 2020 were retrospectively included. EBI was categorized based on intracranial blood burden, persistent loss of consciousness, and SEBES (Subarachnoid Hemorrhage Early Brain Edema Score) score on the third day after ictus. The draining CSF and vital signs such as ICP and CPP were documented daily. RESULTS: 90 out of 324 eligible aSAH patients (28%) were included. The mean age was 54.2 ± 11.9 years. DHC was performed in 24% (22/90) of patients. Mean CSF drainage within 72 h after ictus was 168.5 ± 78.5 ml. A higher CSF drainage within 72 h after ictus correlated with a less severe EBI and a less frequent need for DHC (r=-0.33, p = 0.001) and with a higher mean CPP on day 3 after ictus (r = 0.2351, p = 0.02). CONCLUSION: A more pronounced CSF drainage in the first 3 days of aSAH was associated with higher CPP and a less severe course of EBI and required less frequently a DHC. These results support the hypothesis that an early and pronounced CSF drainage may facilitate blood clearance and positively influence the course of EBI.


Subject(s)
Aneurysm, Ruptured , Drainage , Subarachnoid Hemorrhage , Humans , Middle Aged , Male , Subarachnoid Hemorrhage/surgery , Subarachnoid Hemorrhage/complications , Female , Drainage/methods , Retrospective Studies , Adult , Aneurysm, Ruptured/surgery , Aneurysm, Ruptured/complications , Aged , Decompressive Craniectomy/methods , Brain Injuries , Brain Edema/etiology , Brain Edema/cerebrospinal fluid , Brain Edema/surgery , Cerebrospinal Fluid , Intracranial Hypertension/etiology , Intracranial Hypertension/surgery , Intracranial Hypertension/cerebrospinal fluid , Intracranial Aneurysm/surgery , Intracranial Aneurysm/complications
20.
J Neuroinflammation ; 21(1): 140, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38807233

ABSTRACT

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.


Subject(s)
Brain Edema , Cerebral Hemorrhage , Disease Progression , Sequence Analysis, RNA , Single-Cell Analysis , Humans , Brain Edema/immunology , Brain Edema/pathology , Brain Edema/genetics , Brain Edema/metabolism , Brain Edema/etiology , Cerebral Hemorrhage/immunology , Cerebral Hemorrhage/pathology , Cerebral Hemorrhage/genetics , Male , Female , Middle Aged , Sequence Analysis, RNA/methods , Aged , Hematoma/pathology , Hematoma/immunology , Hematoma/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...