Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 181
Filter
1.
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
2.
BMC Cardiovasc Disord ; 24(1): 266, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773462

ABSTRACT

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.


Subject(s)
Cardiopulmonary Bypass , Disease Models, Animal , Glycogen Synthase Kinase 3 beta , Proto-Oncogene Proteins c-akt , Pyroptosis , Rats, Sprague-Dawley , Signal Transduction , Animals , Cardiopulmonary Bypass/adverse effects , Glycogen Synthase Kinase 3 beta/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyroptosis/drug effects , Male , Neurons/drug effects , Neurons/pathology , Neurons/metabolism , Neurons/enzymology , Neuroprotective Agents/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Brain Edema/prevention & control , Brain Edema/metabolism , Brain Edema/enzymology , Brain Edema/pathology , Anti-Inflammatory Agents/pharmacology , Rats , S100 Calcium Binding Protein beta Subunit/metabolism , Inflammation Mediators/metabolism
3.
Toxicol Appl Pharmacol ; 430: 115728, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34560092

ABSTRACT

1,2-Dichloroethane (1,2-DCE) is a pervasive environmental pollutant found in ambient and residential air, as well as ground and drinking water. Overexposure to it results in cortex edema, in both animals and humans. 1,2-DCE induces apoptosis in the cerebellum, liver and testes. This promotes the hypothesis that 1,2-DCE may induce apoptosis in the cortex as brain edema progresses. To validate our hypothesis, 40 NIH male mice were exposed to 0, 100, 350, 700 mg/m3 1,2-DCE by whole-body dynamic inhalation for 28 consecutive days. MicroRNA (miRNA) and mRNA microarray combined with TdT-mediated dUTP nick-end labeling, flow cytometry, and mitochondrial membrane potential (mtΔΨ) measurement were applied to identify the cortex apoptosis pathways' specific responses to 1,2-DCE, in vitro and in vivo. The results showed that 1,2-DCE caused brain edema and increased apoptosis in the mouse cortexes. We confirmed that 1,2-DCE induced increased apoptosis via mitochondrial pathway, both in vitro and in vivo, as evidenced by increased Caspase-3, cleaved Caspase-3, Cytochrome c and Bax expression, and decreased Bcl-2 expression. Additionally, mtΔΨ decreased after 1,2-DCE treatment in vitro. 1,2-DCE exposure increased miR-182-5p and decreased phospholipase D1 (PLD1) in the cerebral cortex of mice. MiR-182-5p overexpression and PLD1 inhibition reduced mtΔΨ and increased astrocyte apoptosis, yet miR-182-5p inhibition alleviated the 1,2-DCE-induced PLD1 down-regulation and the increased apoptosis. Finally, PLD1 was confirmed to be a target of miR-182-5p by luciferase assay. Taken together, our findings indicate that 1,2-DCE exposure induces apoptosis in the cortex via a mitochondria-dependent pathway. This pathway is regulated by a miR-182-5p⊣PLD1 axie.


Subject(s)
Apoptosis/drug effects , Brain Edema/chemically induced , Cerebral Cortex/drug effects , Environmental Pollutants/toxicity , Ethylene Dichlorides/toxicity , MicroRNAs/metabolism , Mitochondria/drug effects , Phospholipase D/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Brain Edema/enzymology , Brain Edema/genetics , Brain Edema/pathology , Cell Line , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Disease Progression , Male , Membrane Potential, Mitochondrial/drug effects , Mice , MicroRNAs/genetics , Mitochondria/enzymology , Mitochondria/genetics , Mitochondria/pathology , Phospholipase D/genetics , Signal Transduction
4.
Eur J Pharmacol ; 893: 173811, 2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33345851

ABSTRACT

Anti-inflammatory therapy for early brain injury after subarachnoid hemorrhage is a promising treatment for improving the prognosis. HMGB1 is the initiator of early inflammation after subarachnoid hemorrhage. Oleanolic acid is a natural pentacyclic triterpenoid compound with strong anti-inflammatory activity. It can relieve early brain injury in subarachnoid hemorrhage rats, but its mechanism is not very clear. Here, we study the potential mechanism of Oleanolic acid in the treatment of subarachnoid hemorrhage. First, we demonstrated that oleanolic acid alleviated early brain injury after subarachnoid hemorrhage, including improvement of grading score, neurological score, brain edema and permeability of brain blood barrier. Then we found that oleanolic acid could inhibit the transfer of HMGB1 from nucleus to cytoplasm and reduce the level of serum HMGB1. Furthermore, we found that oleanolic acid decreased the acetylation level of HMGB1 by increasing SIRT1 expression rather than by inhibiting JAK/STAT3 pathway. SIRT1 inhibitor sirtinol eliminated all beneficial effects of oleanolic acid on subarachnoid hemorrhage, which indicated that oleanolic acid inhibited the acetylation of HMGB1 by up regulating SIRT1. In addition, oleanolic acid treatment also reduced the levels of TLR4 and apoptosis related factors and reduced neuronal apoptosis after subarachnoid hemorrhage. In summary, our findings suggest that oleanolic acid may activate SIRT1 by acting as an activator of SIRT1, thereby reducing the acetylation of HMGB1, thus playing an anti-inflammatory role to alleviate early brain injury after subarachnoid hemorrhage.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Brain/drug effects , HMGB1 Protein/metabolism , Neuroprotective Agents/pharmacology , Oleanolic Acid/pharmacology , Sirtuin 1/metabolism , Subarachnoid Hemorrhage/drug therapy , Acetylation , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/enzymology , Brain/pathology , Brain Edema/drug therapy , Brain Edema/enzymology , Brain Edema/pathology , Capillary Permeability/drug effects , Disease Models, Animal , HMGB1 Protein/genetics , Male , Protein Processing, Post-Translational , Rats, Sprague-Dawley , Signal Transduction , Sirtuin 1/genetics , Subarachnoid Hemorrhage/enzymology , Subarachnoid Hemorrhage/pathology
5.
Front Neuroendocrinol ; 56: 100816, 2020 01.
Article in English | MEDLINE | ID: mdl-31786088

ABSTRACT

Aromatase is the requisite and limiting enzyme in the production of estrogens from androgens. Estrogens synthesized centrally have more recently emerged as potent neuroprotectants in the vertebrate brain. Studies in rodents and songbirds have identified key mechanisms that underlie both; the injury-dependent induction of central aromatization, and the protective effects of centrally synthesized estrogens. Injury-induced aromatase expression in astrocytes occurs following a broad range of traumatic brain damage including excitotoxic, penetrating, and concussive injury. Responses to neural insult such as edema and inflammation involve signaling pathways the components of which are excellent candidates as inducers of this astrocytic response. Finally, estradiol from astrocytes exerts a paracrine neuroprotective influence via the potent inhibition of inflammatory pathways. Taken together, these data suggest a novel role for neural aromatization as a protective mechanism against the threat of inflammation and suggests that central estrogen provision is a wide-ranging neuroprotectant in the vertebrate brain.


Subject(s)
Aromatase/metabolism , Brain Injuries/enzymology , Brain/enzymology , Animals , Aromatase/genetics , Astrocytes/enzymology , Brain Edema/enzymology , Estradiol/physiology , Female , Humans , Inflammation/enzymology , Male , Nerve Degeneration/enzymology , Neuroprotection/physiology , Songbirds/physiology , Up-Regulation
6.
J Stroke Cerebrovasc Dis ; 28(12): 104470, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31680031

ABSTRACT

BACKGROUND: The nitric oxide (NO)-producing activity of endothelial nitric oxide synthase (eNOS) plays a significant role in maintaining endothelial function and protecting against the stroke injury. However, the activity of the eNOS enzyme and the metabolism of major NO metabolite S-nitrosoglutathione (GSNO) are dysregulated after stroke, causing endothelial dysfunction. We investigated whether an administration of exogenous of GSNO or enhancing the level of endogenous GSNO protects against neurovascular injury in wild-type (WT) and eNOS-null (endothelial dysfunction) mouse models of cerebral ischemia-reperfusion (IR). METHODS: Transient cerebral ischemic injury was induced by middle cerebral artery occlusion (MCAO) for 60 minutes in male adult WT and eNOS null mice. GSNO (0.1 mg/kg body weight, intravenously) or N6022 (GSNO reductase inhibitor, 5.0 mg/kg body weight, intravenously) was administered 30 minutes before MCAO in preinjury and at the reperfusion in postinjury studies. Brain infarctions, edema, and neurobehavioral functions were evaluated at 24 hours after the reperfusion. RESULTS: eNOS-null mice had a higher degree (P< .05) of injury than WT. Pre- or postinjury treatment with either GSNO or N6022 significantly reduced infarct volume, improved neurological and sensorimotor function in both WT and eNOS-null mice. CONCLUSION: Reduced brain infarctions and edema, and improved neurobehavioral functions by pre- or postinjury GSNO treatment of eNOS knock out mice indicate that GSNO can attenuate IR injury, likely by mimicking the eNOS-derived NO-dependent anti-ischemic and anti-inflammatory functions. Neurovascular protection by GSNO/N6022 in both pre- and postischemic injury groups support GSNO as a promising drug candidate for the prevention and treatment of stroke injury.


Subject(s)
Alcohol Dehydrogenase/antagonists & inhibitors , Benzamides/pharmacology , Brain/drug effects , Enzyme Inhibitors/pharmacology , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Pyrroles/pharmacology , S-Nitrosoglutathione/pharmacology , Alcohol Dehydrogenase/metabolism , Animals , Behavior, Animal/drug effects , Brain/embryology , Brain/pathology , Brain Edema/enzymology , Brain Edema/pathology , Brain Edema/prevention & control , Disease Models, Animal , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type III/deficiency , Nitric Oxide Synthase Type III/genetics
7.
J Stroke Cerebrovasc Dis ; 28(12): 104375, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31590996

ABSTRACT

BACKGROUND: Our previous study showed that propofol, one of the widely used anesthetic agents, can attenuate subarachnoid hemorrhage (SAH)-induced early brain injury (EBI) via inhibiting inflammatory and oxidative reaction. However, it is perplexing whether propofol attenuates inflammatory and oxidative reaction through modulating PI3K/Akt pathway. The present study investigated whether PI3K/Akt pathway is involved in propofol's anti-inflammation, antioxidation, and neuroprotection against SAH-induced EBI. MATERIALS AND METHODS: Adult Sprague-Dawley rats underwent SAH and received treatment with propofol or vehicle after 2 and 12 hours of SAH. LY294002 was injected intracerebroventricularly to selectively inhibit PI3K/Akt signaling. Mortality, SAH grading, neurological scores, brain water content, evans blue extravasation, myeloperoxidase, malondialdehyde, superoxide dismutase, and glutathione peroxidase were measured 24 hours after SAH. Immunoreactivity of p-Akt, t-Akt, nuclear factor- kappa B (NF-κB) p65, nuclear factor erythroid-related factor 2 (Nrf2), NAD(P)H:quinone oxidoreductase (NQO1), and cyclooxygenase-2 (COX-2) in rat brain was determined by western blot. Tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß) in rat brain were examined by ELISA. RESULTS: Propofol significantly reduces neurological dysfunction, BBB permeability, brain edema, inflammation, and oxidative stress, all of which were reversed by LY294002. Propofol significantly upregulates the immunoreactivity of p-Akt, Nrf2, and NQO1, all of which were abolished by LY294002. Propofol significantly downregulates the overexpression of NF-κB p65, COX-2, TNF-α, and IL-1ß, all of which were inhibited by LY294002. CONCLUSION: These results suggest that propofol attenuates SAH-induced EBI by inhibiting inflammatory reaction and oxidative stress, which might be associated with the activation of PI3K/Akt signaling pathway.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Brain Edema/prevention & control , Brain/drug effects , Encephalitis/prevention & control , Neuroprotective Agents/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Propofol/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Subarachnoid Hemorrhage/drug therapy , Animals , Brain/enzymology , Brain/pathology , Brain Edema/enzymology , Brain Edema/pathology , Cyclooxygenase 2/metabolism , Disease Models, Animal , Encephalitis/enzymology , Encephalitis/pathology , Interleukin-1beta/metabolism , Male , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Phosphorylation , Rats, Sprague-Dawley , Signal Transduction , Subarachnoid Hemorrhage/enzymology , Subarachnoid Hemorrhage/pathology , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
J Am Heart Assoc ; 8(9): e011801, 2019 05 07.
Article in English | MEDLINE | ID: mdl-31030600

ABSTRACT

Background Systemic innate immune priming is a recognized sequela of post-ischemic neuroinflammation and contributor to delayed neurodegeneration. Given mounting evidence linking acute stroke with reactive lung inflammation, we asked whether enhanced expression of the endogenous antioxidant extracellular superoxide dismutase 3 (SOD3) produced by alveolar type II pneumocytes would protect the lung from transient global cerebral ischemia and the brain from the delayed effects of ischemia-reperfusion. Methods and Results Following 15 minutes of global cerebral ischemia or sham conditions, transgenic SOD3 and wild-type mice were followed daily for changes in weight, core temperature, and neurological function. Three days after reperfusion, arterial and venous samples were collected for complete blood counts, flow cytometry, and SOD3 protein blotting, and immunohistochemistry was performed on lung and brain tissue to assess tissue injury, blood-brain barrier permeability, and neutrophil transmigration. Relative to ischemic controls, transgenic SOD3 mice performed better on functional testing and exhibited reduced peripheral neutrophil activation, lung inflammation, and blood-brain barrier leak. Once released from the lung, SOD3 was predominantly not cell associated and depleted in the venous phase of circulation. Conclusions In addition to reducing the local inflammatory response to cerebral ischemia, targeted enrichment of SOD3 within the lung confers distal neuroprotection against ischemia-reperfusion injury. These data suggest that therapies geared toward enhancing adaptive lung-neurovascular coupling may improve outcomes following acute stroke and cardiac arrest.


Subject(s)
Brain Ischemia/enzymology , Brain/enzymology , Lung/enzymology , Neurovascular Coupling , Pneumonia/prevention & control , Reperfusion Injury/prevention & control , Superoxide Dismutase/metabolism , Animals , Blood-Brain Barrier/enzymology , Blood-Brain Barrier/immunology , Blood-Brain Barrier/pathology , Brain/immunology , Brain/pathology , Brain Edema/enzymology , Brain Edema/pathology , Brain Edema/prevention & control , Brain Ischemia/immunology , Brain Ischemia/pathology , Capillary Permeability , Disease Models, Animal , Humans , Immunity, Innate , Lung/immunology , Male , Mice, Inbred C57BL , Mice, Transgenic , Neutrophil Infiltration , Pneumonia/enzymology , Pneumonia/immunology , Pneumonia/pathology , Pulmonary Edema/enzymology , Pulmonary Edema/pathology , Pulmonary Edema/prevention & control , Reperfusion Injury/enzymology , Reperfusion Injury/immunology , Reperfusion Injury/pathology , Superoxide Dismutase/genetics
9.
Radiology ; 290(2): 359-367, 2019 02.
Article in English | MEDLINE | ID: mdl-30615566

ABSTRACT

Purpose To investigate the association of inflammation and brain edema in a cerebral malaria (CM) mouse model with a combination of bis-5-hydroxy-tryptamide-diethylenetriaminepentaacetate gadolinium, referred to as MPO-Gd, and cross-linked iron oxide nanoparticle (CLIO-NP) imaging. Materials and Methods Female wild-type (n = 23) and myeloperoxidase (MPO) knock-out (n = 5) mice were infected with the Plasmodium berghei ANKA strain from May 2016 to July 2018. Seven healthy mice served as control animals. At a Rapid Murine Coma and Behavioral Scale (RMCBS) score of less than 15, mice underwent MRI at 9.4 T and received gadodiamide, MPO-Gd, or CLIO-NPs. T1-weighted MRI was used to assess MPO activity, and T2*-weighted MRI was used to track CLIO-NPs. Immunofluorescent staining and flow cytometric analyses characterized CLIO-NPs, MPO, endothelial cells, and leukocytes. An unpaired, two-tailed Student t test was used to compare groups; Spearman correlation analysis was used to determine the relationship of imaging parameters to clinical severity. Results MPO-Gd enhancement occurred in inflammatory CM hotspots (olfactory bulb > rostral migratory stream > brainstem > cortex, P < .05 for all regions compared with control mice; mean olfactory bulb signal intensity ratio: 1.40 ± 0.07 vs 0.96 ± 0.01, P < .01). The enhancement was reduced in MPO knockout mice (mean signal intensity ratio at 60 minutes: 1.13 ± 0.04 vs 1.40 ± 0.07 in CM, P < .05). Blood-brain barrier compromise was suggested by parenchymal gadolinium enhancement, leukocyte recruitment, and endothelial activation. CLIO-NPs accumulated mainly intravascularly and at the vascular endothelium. CLIO-NPs were also found in the choroid plexus, indicating inflammation of the ventricular system. Blood-cerebrospinal fluid barrier breakdown showed correlation with brain swelling (r2: 0.55, P < .01) and RMCBS score (r2: 0.75, P < .001). Conclusion Iron oxide nanoparticle imaging showed strong inflammatory involvement of the microvasculature in a murine model of cerebral malaria. Furthermore, bis-5-hydroxy-tryptamide-diethylenetriaminepentaacetate gadolinium imaging depicted parenchymal and intraventricular inflammation. This combined molecular imaging approach links vascular inflammation to breakdown of the blood-brain barrier and blood-cerebrospinal fluid barrier that correlate with global brain edema and disease severity. © RSNA, 2018 Online supplemental material is available for this article. See also the editorial by Kiessling in this issue.


Subject(s)
Brain Edema , Encephalitis , Magnetic Resonance Imaging/methods , Magnetite Nanoparticles/chemistry , Malaria, Cerebral , Peroxidase/metabolism , Animals , Brain/diagnostic imaging , Brain/enzymology , Brain/pathology , Brain Edema/diagnostic imaging , Brain Edema/enzymology , Brain Edema/parasitology , Brain Edema/pathology , Disease Models, Animal , Encephalitis/diagnostic imaging , Encephalitis/enzymology , Encephalitis/parasitology , Encephalitis/pathology , Female , Malaria, Cerebral/complications , Malaria, Cerebral/diagnostic imaging , Malaria, Cerebral/enzymology , Malaria, Cerebral/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout
10.
J Cell Physiol ; 234(4): 5304-5318, 2019 04.
Article in English | MEDLINE | ID: mdl-30216439

ABSTRACT

Inflammation and apoptosis are two key factors contributing to secondary brain injury after intracerebral hemorrhage (ICH). In the present study, we explored the neuroprotective role of methylene blue (MB) in ICH rats and studied the potential mechanisms involved. Rats were subjected to local injection of collagenase IV in the striatum or sham surgery. We observed that MB treatment could exert a neuroprotective effect on ICH by promoting neurological scores, decreasing the brain water content, alleviating brain-blood barrier disruption, and improving the histological damages in the perihematomal areas. Furthermore, we demonstrated that the various mechanisms underlying MB's neuroprotective effects linked to inhibited apoptosis and inhibited neuroinflammation. In addition, wortmannin, a selective inhibitor of phosphoinositide 3-kinase (PI3K), could reverse the antiapoptotic and anti-inflammatory effects of MB, which suggested that the PI3K-Akt pathway played an important role. In conclusion, these data suggested that MB could inhibit apoptosis and ameliorate neuroinflammation after ICH, and its neuroprotective effects might be exerted via the activation of the PI3K/Akt/GSK3ß pathway.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Brain/drug effects , Cerebral Hemorrhage/drug therapy , Encephalitis/prevention & control , Glycogen Synthase Kinase 3 beta/metabolism , Methylene Blue/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis/drug effects , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/enzymology , Blood-Brain Barrier/pathology , Brain/enzymology , Brain/pathology , Brain Edema/enzymology , Brain Edema/pathology , Brain Edema/prevention & control , Capillary Permeability/drug effects , Cerebral Hemorrhage/complications , Cerebral Hemorrhage/enzymology , Cerebral Hemorrhage/pathology , Cytokines/metabolism , Disease Models, Animal , Encephalitis/enzymology , Encephalitis/etiology , Encephalitis/pathology , Male , Microglia/drug effects , Microglia/enzymology , Microglia/pathology , Neurons/enzymology , Neurons/pathology , Neutrophil Infiltration/drug effects , Rats, Sprague-Dawley , Signal Transduction
11.
Drug Des Devel Ther ; 12: 2497-2508, 2018.
Article in English | MEDLINE | ID: mdl-30127597

ABSTRACT

BACKGROUND: The neuroprotective effects of Baicalin have been confirmed in several central nervous system (CNS) diseases. However, its possible effect on traumatic brain injury (TBI) model is still not clear. The present study is aimed to investigate the role and the underling mechanisms of 7-D-glucuronic acid-5,6-dihydroxyflavone (Baicalin) on TBI model. METHODS: The weight-drop model of TBI in Institute of Cancer Research mice was treated with Baicalin intraperitoneally at 30 minutes after TBI. LY294002 (LY) (a commonly used PI3K/Akt pathway inhibitor) was injected into the left ventricle at 30 minutes before TBI. All mice were euthanized at 24 hours after TBI to collect the brain tissue for a series of tests except for neurological function, which was measured at 2 hours and 1 and 3 days post-TBI. RESULTS: Baicalin administration significantly improved neurobehavioral function, alleviated brain edema, and reduced apoptosis-positive cells by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay accompanied with the upregulation of B-cell lymphoma 2 (Bcl-2) and downregulation of Bcl-2-associated X protein (Bax) and cleaved-caspase 3 by Western blot. Besides, TBI-induced oxidant stress status was also restored in the Baicalin group by measuring malondialdehyde (MDA) content, glutathione peroxidase (GPx), and superoxide dismutase (SOD) levels in the injured brain cortex. Furthermore, translocation of Nrf2 to the nucleus was dramatically enhanced by Baicalin verified by immunofluorescence and Western blot analyses. Accordingly, its downstream antioxidative enzymes nicotinamide adenine dinucleotide phosphate:quinine oxidoreductase 1 (NQO-1) and heme oxygenase 1 (HO-1) were also activated by Baicalin confirmed by quantitative reverse transcription polymerase chain reaction (RT-qPCR) and Western blot. However, cotreatment with Baicalin and LY could partly abolish Baicalin-induced activation of Nrf2 and its neuroprotective effects in TBI. CONCLUSION: This study demonstrates that Baicalin provides a neuroprotective effect in TBI mice model via activating the Akt/Nrf2 pathway.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Brain/drug effects , Flavonoids/pharmacology , NF-E2-Related Factor 2/metabolism , Neurons/drug effects , Neuroprotective Agents/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Behavior, Animal/drug effects , Brain/enzymology , Brain/pathology , Brain/physiopathology , Brain Edema/enzymology , Brain Edema/pathology , Brain Edema/prevention & control , Brain Injuries, Traumatic/enzymology , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/psychology , Disease Models, Animal , Male , Mice, Inbred ICR , Neurons/enzymology , Neurons/pathology , Oxidative Stress/drug effects , Signal Transduction/drug effects
12.
J Stroke Cerebrovasc Dis ; 27(11): 3053-3065, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30093209

ABSTRACT

BACKGROUND: The present study aimed to examine the protective role of fullerenol nanoparticles against blood-brain barrier (BBB) interruption and brain edema during cerebral ischemia-reperfusion injury probably by reduction of interleukin-6 (IL-6) and matrix metalloproteinase-9 (MMP-9) transcription. METHODS: The male Wistar rats (weighting 280-320 g) were randomly assigned into four groups as follows: sham, control ischemic, pretreated ischemic, and posttreated ischemic groups. Cerebral ischemia-reperfusion (IR) injury was performed by occlusion of middle cerebral artery (MCA) for 90 minutes followed by twenty-four hours reperfusion. Rats were administered fullerenol 5mg/kg, intraperitoneally, 30 minutes before induction of IR in pretreated ischemic group and immediately after termination of MCA occlusion in posttreated ischemic group. After twenty-four hours reperfusion, the method of Evans blue dye extravasation (EBE) and RT-PCR were used for determination of BBB permeability and mRNA expression levels of MMP-9 and IL-6, respectively. Neuronal deficit score (NDS) and edema of the ischemic hemispheres were also evaluated. RESULTS: MCA occlusion increased NDS in control ischemic rats (3.16 ± 0.16) with concomitant increase in EBE (15.30 ± 3.98µg/g) and edema (3.53 ± 0.50%). Fullerenol in both pretreated and posttreated ischemic groups reduced NDS (36% and 68%, respectively), EBE (89% and 91%, respectively) and edema (53% and 81%, respectively). Although MCA occlusion increased the mRNA expression levels of MMP-9 and IL-6 in ischemic hemispheres, fullerenol in both treatment groups noticeably decreased the mRNA expression levels of these genes. CONCLUSION: In conclusion, fullerenol nanoparticles can protect BBB integrity and attenuate brain edema after cerebral ischemia-reperfusion injury possibly by reduction of IL-6 and MMP-9 transcription.


Subject(s)
Blood-Brain Barrier/drug effects , Brain Edema/prevention & control , Fullerenes/pharmacology , Infarction, Middle Cerebral Artery/drug therapy , Interleukin-6/metabolism , Matrix Metalloproteinase 9/metabolism , Nanoparticles , Neuroprotective Agents/pharmacology , Reperfusion Injury/prevention & control , Transcription, Genetic/drug effects , Animals , Blood-Brain Barrier/enzymology , Blood-Brain Barrier/pathology , Brain Edema/enzymology , Brain Edema/genetics , Brain Edema/pathology , Capillary Permeability/drug effects , Disease Models, Animal , Down-Regulation , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Interleukin-6/genetics , Male , Matrix Metalloproteinase 9/genetics , Rats, Wistar , Reperfusion Injury/enzymology , Reperfusion Injury/genetics , Reperfusion Injury/pathology
13.
Neurotoxicology ; 69: 296-306, 2018 12.
Article in English | MEDLINE | ID: mdl-30081051

ABSTRACT

Accumulated data have revealed that subacute poisoning of 1,2-dichloroethane (1,2-DCE), an industrial solvent used in some countries can cause encephalopathy, in which brain edema is the main pathological change. However, the underlying mechanisms are unclear. In the present study, we hypothesized that the p38 MAPK (p38) signaling pathway could be activated in 1,2-DCE-intoxicated mice, which in turn stimulates transcription factors, such as nuclear factor-κB (NF-κB) and activator protein-1 (AP-1), and then enhances the expression of proinflammatory factors, including matrix metalloproteinase-9 (MMP-9), finally leading to blood-brain barrier (BBB) disruption and brain edema formation. Our results revealed that brain water content and BBB permeability increased significantly in the intoxicated mice. Meanwhile, the levels of phosphorylated p38 (p-p38) and inhibitory κBα (p-IκB), as well as the expression levels of MMP-9, c-jun, c-fos, and p65, also increased markedly in the brains of intoxicated mice. Conversely, the protein levels of ZO-1, occludin and claudin-5 in these mice decreased markedly, but their JAM-1 protein levels increased dramatically. Our results revealed that p-p38 levels in the brains of intoxicated mice were suppressed by pretreatment with a p38 inhibitor. In response to suppressed p-p38 levels, the brain water contents and DNA binding activities of NF-κB and AP-1, as well as the expression levels of MMP-9, c-jun, c-fos, p65, p-IκB and JAM-1, decreased, whereas the protein levels of ZO-1, occludin and claudin-5 increased markedly. Taken together, our findings indicated that the p38 signaling pathway might be activated and involved in the course of brain edema in 1,2-DCE-intoxicated mice.


Subject(s)
Brain Edema/chemically induced , Brain Edema/enzymology , Ethylene Dichlorides/toxicity , Matrix Metalloproteinase 9/biosynthesis , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Female , Gene Expression Regulation, Enzymologic , Matrix Metalloproteinase 9/genetics , Mice , Signal Transduction/physiology
14.
Biomed Pharmacother ; 103: 1720-1726, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29864962

ABSTRACT

INTRODUCTION: Stroke is a prevalent disorder which is associated with several complications including inflammation. JZL-184 (JZL) inhibits arachidonic acid (AA) production and consequently results in two-arachidonoylglycerol (2-AG) accumulation. Both reduced production of AA metabolic products and increased 2-AG, the agonist of type 1 cannabinoid receptor (CB1), can result in reduced inflammation. In this study, we investigated the mechanisms of JZL in the improvement of stroke complications in mouse permanent cerebral ischemia (PPMCAO) model using AM251, the antagonist of CB1. MATERIAL AND METHODS: PMCAO mice were divided into six groups including intact, controls, vehicle, JZL, AM251 and JZL plus AM251 administrated groups. Brain infarction and edema, brain levels of matrix metalloperoteinase-9 (MMP9), interleukin (IL)-10 and tumor necrosis factor-α (TNF-α) and behavioral functions have been examined in all groups. RESULTS: The results showed that JZL lowered brain infarction, neurological disorders, TNF-α and MMP9 more effectively than JZL plus AM251. JZL and JZL plus AM251 reduced brain edema and increased brain IL-10. JZL, AM251 and JZL plus AM251 improve behavioral functions. DISCUSSION: JZL reduces brain infarction and brain pro-inflammatory molecules in CB1 pathway dependent manner. JZL also reduces brain edema and increased IL-10 in CB1 pathways or decreased AA metabolites. Further, AM251 improves behavioral functions via unknown mechanisms.


Subject(s)
Benzodioxoles/pharmacology , Cannabinoids/metabolism , Down-Regulation/drug effects , Enzyme Inhibitors/pharmacology , Inflammation/pathology , Monoacylglycerol Lipases/antagonists & inhibitors , Piperidines/pharmacology , Signal Transduction/drug effects , Animals , Benzodioxoles/therapeutic use , Brain Edema/complications , Brain Edema/drug therapy , Brain Edema/enzymology , Brain Edema/pathology , Brain Infarction/complications , Brain Infarction/drug therapy , Brain Infarction/enzymology , Brain Infarction/pathology , Inflammation/complications , Inflammation/drug therapy , Interleukin-10/metabolism , Male , Matrix Metalloproteinase 9/metabolism , Mice , Monoacylglycerol Lipases/metabolism , Piperidines/therapeutic use , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Tumor Necrosis Factor-alpha/metabolism
16.
Am J Transplant ; 17(5): 1405-1408, 2017 May.
Article in English | MEDLINE | ID: mdl-27997078

ABSTRACT

Ornithine transcarbamylase deficiency represents the most common inherited defect of the urea cycle. This enzyme, predominantly found in the liver, plays a crucial role in recycling free ammonia, with deficiencies often leading to fatal complications. Here, we present the case of a 63-year-old man with alcoholic cirrhosis who underwent orthotopic liver transplantation, gradual worsening of his mental status, and progressive elevation of ammonia levels. Liver allograft function was deemed normal, raising concern for a donor-derived metabolic disorder of the urea cycle. Evaluation of the donor patient's blood revealed that the donor was heterozygous for the OTC gene. Posttransplantation changes in mental status should prompt a clinician to consider the most likely causes; however, once these have been ruled out, it is important to consider the less common causes of metabolic derangements. The rarity of these disorders makes expertise of diagnosis, standardization of evaluation, and treatment strategies challenging.


Subject(s)
Brain Edema/etiology , Hyperammonemia/etiology , Liver Transplantation/adverse effects , Ornithine Carbamoyltransferase Deficiency Disease/complications , Ornithine Carbamoyltransferase/metabolism , Tissue Donors , Brain Edema/enzymology , Humans , Hyperammonemia/enzymology , Male , Middle Aged , Prognosis , Transplantation, Homologous
17.
Neurosci Lett ; 627: 71-6, 2016 08 03.
Article in English | MEDLINE | ID: mdl-27241721

ABSTRACT

Herpes simplex encephalitis(HSE) is the most common and serious viral encephalitis in humans. There is a lack of effective medication to date for HSE. A better understanding of the mediators of tissue damage is essential for finding new targets for therapeutic intervention. In this project, we explored the effect of cyclin-dependent kinases inhibitor olomoucine treatment on experimental HSE mice. The following results were obtained: (1) olomoucine increased survival in HSE mice; (2) olomoucine inhibited microglial activation and reduced HSV-1-induced cytokines release; (3) olomoucine prevented neural cells apoptosis and attenuated brain tissue pathological changes following HSV-1 infection; (4) olomoucine reduced brain edema and improved neurological function in HSE. Overall, olomoucine can induce a blunted inflammatory response, maintain the blood vessel wall intact, improve neurological function and increase survival in HSE mice.


Subject(s)
Apoptosis/drug effects , Brain/drug effects , Brain/virology , Cyclin-Dependent Kinases/antagonists & inhibitors , Encephalitis, Herpes Simplex/enzymology , Enzyme Inhibitors/administration & dosage , Kinetin/administration & dosage , Animals , Brain/pathology , Brain Edema/enzymology , Brain Edema/prevention & control , Encephalitis, Herpes Simplex/pathology , Female , Herpesvirus 1, Human/pathogenicity , Kaplan-Meier Estimate , Mice , Mice, Inbred BALB C , Microglia/drug effects , Microglia/enzymology , Viral Envelope Proteins/metabolism
18.
Crit Care Med ; 44(9): e846-53, 2016 09.
Article in English | MEDLINE | ID: mdl-27071071

ABSTRACT

OBJECTIVE: Diabetic ketoacidosis in children is associated with vasogenic cerebral edema, possibly due to the release of destructive polymorphonuclear neutrophil azurophilic enzymes. Our objectives were to measure plasma azurophilic enzyme levels in children with diabetic ketoacidosis, to correlate plasma azurophilic enzyme levels with diabetic ketoacidosis severity, and to determine whether azurophilic enzymes disrupt the blood-brain barrier in vitro. DESIGN: Prospective clinical and laboratory study. SETTING: The Children's Hospital, London Health Sciences Centre. SUBJECTS: Pediatric type 1 diabetes patients; acute diabetic ketoacidosis or age-/sex-matched insulin-controlled. MEASUREMENTS AND MAIN RESULTS: Acute diabetic ketoacidosis in children was associated with elevated polymorphonuclear neutrophils. Plasma azurophilic enzymes were elevated in diabetic ketoacidosis patients, including human leukocyte elastase (p < 0.001), proteinase-3 (p < 0.01), and myeloperoxidase (p < 0.001). A leukocyte origin of human leukocyte elastase and proteinase-3 in diabetic ketoacidosis was confirmed with buffy coat quantitative real-time polymerase chain reaction (p < 0.01). Of the three azurophilic enzymes elevated, only proteinase-3 levels correlated with diabetic ketoacidosis severity (p = 0.002). Recombinant proteinase-3 applied to human brain microvascular endothelial cells degraded both the tight junction protein occludin (p < 0.05) and the adherens junction protein VE-cadherin (p < 0.05). Permeability of human brain microvascular endothelial cell monolayers was increased by recombinant proteinase-3 application (p = 0.010). CONCLUSIONS: Our results indicate that diabetic ketoacidosis is associated with systemic polymorphonuclear neutrophil activation and degranulation. Of all the polymorphonuclear neutrophil azurophilic enzymes examined, only proteinase-3 correlated with diabetic ketoacidosis severity and potently degraded the blood-brain barrier in vitro. Proteinase-3 might mediate vasogenic edema during diabetic ketoacidosis, and selective proteinase-3 antagonists may offer future vascular- and neuroprotection.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Edema/enzymology , Diabetic Ketoacidosis/enzymology , Leukocyte Elastase/blood , Myeloblastin/blood , Peroxidase/blood , Brain Edema/etiology , Case-Control Studies , Cathepsin G/blood , Cell Culture Techniques , Child , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/enzymology , Diabetic Ketoacidosis/complications , Endothelial Cells/physiology , Female , Humans , Male
19.
Mol Neurobiol ; 53(2): 1310-1321, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25631713

ABSTRACT

The development and/or progression of perihematomal edema (PHE) in patients with acute spontaneous intracerebral hemorrhage (ICH) vary substantially with different individuals. Although hematoma volume is a useful indicator for predicting PHE, its predictive power was not good at the early stage of ICH. Better predictors are urgently needed. In this study, we found that miR-130a was elevated in the serum of ICH patients and was an independent indicator positively associated with PHE volume within the first 3 days after onset. The R (2) was further evaluated when it is used in combination with hematoma mass. Serum miR-130a levels were associated with clinical outcome (National Institute of Health Stroke Scale (NIHSS) scores at day 14 and modified Rankin Scale (mRS) scores at day 90) only in patients with deep hematoma. Moreover, miR-130a was significantly increased in rat serum and perihematomal tissues and was in line with the change in brain edema. MiR-130a inhibitors reduced brain edema, blood-brain barrier (BBB) permeability, and increased neurological deficit scores, and miR-130a mimics increased monolayer permeability. Thrombin-stimulated brain microvascular endothelial cells (BMECs) were a main source of miR-130a under ICH. In the experimental model, the elevated miR-130a level was accompanied by the decreased caveolin-1 and increased matrix metalloproleinase (MMP)-2/9. Meanwhile, caveolin-1 (cav-1) was reduced by miR-130a mimics, accompanied by an increase in MMP-2/9 expression. The upregulated MMP-2/9 was then downregulated by cavtratin, a cav-1 scaffolding domain peptide. This regulation mechanism was authenticated in a thrombin-induced cellular ICH model. Our results suggest that serum miR-130a may serve as a useful early biomarker for monitoring post-ICH PHE and predicting prognosis and may be helpful in the decision-making of individualized therapy.


Subject(s)
Brain Edema/blood , Brain Edema/genetics , Cerebral Hemorrhage/blood , Cerebral Hemorrhage/genetics , MicroRNAs/blood , Acute Disease , Animals , Behavior, Animal , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain/pathology , Brain Edema/complications , Brain Edema/enzymology , Caveolin 1/metabolism , Cerebral Hemorrhage/complications , Cerebral Hemorrhage/enzymology , Demography , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Hematoma/blood , Hematoma/complications , Hematoma/genetics , Humans , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Microvessels/pathology , Middle Aged , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Thrombin/pharmacology , Treatment Outcome , Up-Regulation/genetics
20.
Bull Exp Biol Med ; 160(1): 96-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26608376

ABSTRACT

Adult rats were subjected to 7-day combined stress with stochastic changes of stressors of different modalities (noise, vibration, pulsating bright light) along with mobility restriction and elevated temperature in the chamber during stress exposures (daily 30-min sessions). Circulatory disorders, inhibition of endothelial NO-synthase expression in endothelial cells of the microcirculatory bed, perivascular edema, pronounced degenerative changes, and enhanced expression of inducible NO synthase in CA3 pyramidal neurons in the ventral hippocampus of stressed 12-month-old rats were observed. These findings can attest to the involvement NOdependent mechanisms and different contribution of NO synthase isoforms into the formation of hippocampal neuronal damage.


Subject(s)
CA3 Region, Hippocampal/enzymology , Nerve Tissue Proteins/biosynthesis , Nitric Oxide Synthase Type III/biosynthesis , Nitric Oxide Synthase Type II/biosynthesis , Pyramidal Cells/enzymology , Stress, Physiological , Animals , Animals, Outbred Strains , Brain Edema/enzymology , Brain Edema/etiology , Brain Edema/pathology , CA3 Region, Hippocampal/blood supply , CA3 Region, Hippocampal/ultrastructure , Endothelial Cells/enzymology , Enzyme Induction , Light/adverse effects , Male , Microcirculation , Nerve Degeneration/enzymology , Nerve Degeneration/etiology , Nerve Degeneration/pathology , Nerve Tissue Proteins/genetics , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type III/genetics , Noise/adverse effects , Pyramidal Cells/ultrastructure , Rats , Restraint, Physical/adverse effects , Temperature , Vibration/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL