Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 11.447
Filter
1.
Sci Rep ; 14(1): 15175, 2024 07 02.
Article in English | MEDLINE | ID: mdl-38956251

ABSTRACT

In the current study, we aimed to investigate whether disulfiram (DSF) exerts a neuroprotective role in cerebral ischemiareperfusion (CI-RI) injury by modulating ferredoxin 1 (FDX1) to regulate copper ion (Cu) levels and inhibiting inflammatory responses. To simulate CI-RI, a transient middle cerebral artery occlusion (tMCAO) model in C57/BL6 mice was employed. Mice were administered with or without DSF before and after tMCAO. Changes in infarct volume after tMCAO were observed using TTC staining. Nissl staining and hematoxylin-eosin (he) staining were used to observe the morphological changes of nerve cells at the microscopic level. The inhibitory effect of DSF on initial inflammation was verified by TUNEL assay, apoptosis-related protein detection and iron concentration detection. FDX1 is the main regulatory protein of copper death, and the occurrence of copper death will lead to the increase of HSP70 stress and inflammatory response. Cuproptosis-related proteins and downstream inflammatory factors were detected by western blotting, immunofluorescence staining, and immunohistochemistry. The content of copper ions was detected using a specific kit, while electron microscopy was employed to examine mitochondrial changes. We found that DSF reduced the cerebral infarction volume, regulated the expression of cuproptosis-related proteins, and modulated copper content through down regulation of FDX1 expression. Moreover, DSF inhibited the HSP70/TLR-4/NLRP3 signaling pathway. Collectively, DSF could regulate Cu homeostasis by inhibiting FDX1, acting on the HSP70/TLR4/NLRP3 pathway to alleviate CI/RI. Accordingly, DSF could mitigate inflammatory responses and safeguard mitochondrial integrity, yielding novel therapeutic targets and mechanisms for the clinical management of ischemia-reperfusion injury.


Subject(s)
Copper , Disulfiram , Homeostasis , Inflammation , Mice, Inbred C57BL , Reperfusion Injury , Animals , Reperfusion Injury/metabolism , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , Disulfiram/pharmacology , Mice , Copper/metabolism , Homeostasis/drug effects , Male , Inflammation/metabolism , Inflammation/drug therapy , Inflammation/pathology , Down-Regulation/drug effects , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/drug therapy , Disease Models, Animal , Iron-Sulfur Proteins/metabolism , Brain Ischemia/metabolism , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Apoptosis/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Neuroprotective Agents/pharmacology , Toll-Like Receptor 4/metabolism
2.
Neurotox Res ; 42(4): 35, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39008165

ABSTRACT

This study elucidates the molecular mechanisms by which FABP3 regulates neuronal apoptosis via mitochondrial autophagy in the context of cerebral ischemia-reperfusion (I/R). Employing a transient mouse model of middle cerebral artery occlusion (MCAO) established using the filament method, brain tissue samples were procured from I/R mice. High-throughput transcriptome sequencing on the Illumina CN500 platform was performed to identify differentially expressed mRNAs. Critical genes were selected by intersecting I/R-related genes from the GeneCards database with the differentially expressed mRNAs. The in vivo mechanism was explored by infecting I/R mice with lentivirus. Brain tissue injury, infarct volume ratio in the ischemic penumbra, neurologic deficits, behavioral abilities, neuronal apoptosis, apoptotic factors, inflammatory factors, and lipid peroxidation markers were assessed using H&E staining, TTC staining, Longa scoring, rotation experiments, immunofluorescence staining, and Western blot. For in vitro validation, an OGD/R model was established using primary neuron cells. Cell viability, apoptosis rate, mitochondrial oxidative stress, morphology, autophagosome formation, membrane potential, LC3 protein levels, and colocalization of autophagosomes and mitochondria were evaluated using MTT assay, LDH release assay, flow cytometry, ROS/MDA/GSH-Px measurement, transmission electron microscopy, MitoTracker staining, JC-1 method, Western blot, and immunofluorescence staining. FABP3 was identified as a critical gene in I/R through integrated transcriptome sequencing and bioinformatics analysis. In vivo experiments revealed that FABP3 silencing mitigated brain tissue damage, reduced infarct volume ratio, improved neurologic deficits, restored behavioral abilities, and attenuated neuronal apoptosis, inflammation, and mitochondrial oxidative stress in I/R mice. In vitro experiments demonstrated that FABP3 silencing restored OGD/R cell viability, reduced neuronal apoptosis, and decreased mitochondrial oxidative stress. Moreover, FABP3 induced mitochondrial autophagy through ROS, which was inhibited by the free radical scavenger NAC. Blocking mitochondrial autophagy with sh-ATG5 lentivirus confirmed that FABP3 induces mitochondrial dysfunction and neuronal apoptosis by activating mitochondrial autophagy. In conclusion, FABP3 activates mitochondrial autophagy through ROS, leading to mitochondrial dysfunction and neuronal apoptosis, thereby promoting cerebral ischemia-reperfusion injury.


Subject(s)
Apoptosis , Autophagy , Fatty Acid Binding Protein 3 , Mitochondria , Neurons , Reperfusion Injury , Animals , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Apoptosis/physiology , Autophagy/physiology , Neurons/metabolism , Neurons/pathology , Mice , Mitochondria/metabolism , Male , Fatty Acid Binding Protein 3/metabolism , Fatty Acid Binding Protein 3/genetics , Mice, Inbred C57BL , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/metabolism , Brain Ischemia/metabolism , Brain Ischemia/pathology , Oxidative Stress/physiology
3.
Exp Cell Res ; 440(1): 114127, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38857839

ABSTRACT

CCAAT enhancer binding protein delta (CEBPD) is a transcription factor and plays an important role in apoptosis and oxidative stress, which are the main pathogenesis of ischemic stroke. However, whether CEBPD regulates ischemic stroke through targeting apoptosis and oxidative stress is unclear. Therefore, to answer this question, rat middle cerebral artery occlusion (MCAO) reperfusion model and oxygen-glucose deprivation/reoxygenation (OGD/R) primary cortical neuron were established to mimic ischemic reperfusion injury. We found that CEBPD was upregulated and accompanied with increased neurological deficit scores and infarct size, and decreased neuron in MCAO rats. The siRNA targeted CEBPD inhibited CEBPD expression in rats, and meanwhile lentivirus system was used to blocked CEBPD expression in primary neuron. CEBPD degeneration decreased neurological deficit scores, infarct size and brain water content of MCAO rats. Knockdown of CEBPD enhanced cell viability and reduced apoptosis as well as oxidative stress in vivo and in vitro. CEBPD silencing promoted the translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) to the nucleus and the expression of heme oxygenase 1 (HO-1). Newly, CEBPD facilitated the transcription of cullin 3 (CUL3), which intensified ischemic stroke through Nrf2/HO-1 pathway that was proposed by our team in the past. In conclusion, targeting CEBPD-CUL3-Nrf2/HO-1 axis may be contributed to cerebral ischemia therapy.


Subject(s)
Apoptosis , Heme Oxygenase-1 , Ischemic Stroke , NF-E2-Related Factor 2 , Neurons , Oxidative Stress , Rats, Sprague-Dawley , Animals , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Neurons/metabolism , Neurons/pathology , Ischemic Stroke/metabolism , Ischemic Stroke/pathology , Rats , Male , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , CCAAT-Enhancer-Binding Protein-delta/metabolism , CCAAT-Enhancer-Binding Protein-delta/genetics , Signal Transduction , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Heme Oxygenase (Decyclizing)
4.
J Cell Mol Med ; 28(11): e18366, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38856956

ABSTRACT

Ischemic stroke is one of the main causes of disability and death. However, recanalization of occluded cerebral arteries is effective only within a very narrow time window. Therefore, it is particularly important to find neuroprotective biological targets for cerebral artery recanalization. Here, gene expression profiles of datasets GSE160500 and GSE97537 were downloaded from the GEO database, which were related to ischemic stroke in rats. Olfactory receptor 78 (Olfr78) was screened, and which highly associated with Calcium signalling pathway and MAPK pathway. Interacting protein of Olfr78, Prkaca, was predicted by STRING, and their interaction was validated by Co-IP analysis. Then, a rat model of middle cerebral artery occlusion/reperfusion (MCAO/R) and a neuronal cell model stimulated by oxygen-glucose deprivation/reoxygenation (OGD/R) were constructed, and the results showed that expression of Olfr78 and Prkaca was downregulated in MCAO rats and OGD/R-stimulated neurons. Overexpression of Olfr78 or Prkaca inhibited the secretion of inflammatory factors, Ca2+ overload, and OGD/R-induced neuronal apoptosis. Moreover, Overexpression of Prkaca increased protein levels of cAMP, PKA and phosphorylated p38 in OGD/R-stimulated neurons, while SB203580, a p38 inhibitor, treatment inhibited activation of the cAMP/PKA-MAPK pathway and counteracted the effect of Olfr78 overexpression on improvement of neuronal functions. Meanwhile, overexpression of Olfr78 or Prkaca markedly inhibited neuronal apoptosis and improved brain injury in MCAO/R rats. In conclusion, overexpression of Olfr78 inhibited Ca2+ overload and reduced neuronal apoptosis in MCAO/R rats by promoting Prkaca-mediated activation of the cAMP/PKA-MAPK pathway, thereby improving brain injury in cerebral ischaemia-reperfusion.


Subject(s)
Apoptosis , Cyclic AMP , Rats, Sprague-Dawley , Receptors, Odorant , Reperfusion Injury , Animals , Reperfusion Injury/metabolism , Reperfusion Injury/genetics , Rats , Male , Cyclic AMP/metabolism , Receptors, Odorant/metabolism , Receptors, Odorant/genetics , Brain Ischemia/metabolism , Brain Ischemia/genetics , Brain Ischemia/pathology , MAP Kinase Signaling System/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Brain Injuries/metabolism , Brain Injuries/etiology , Brain Injuries/pathology , Neurons/metabolism , Disease Models, Animal , Infarction, Middle Cerebral Artery/metabolism , Signal Transduction
5.
Sud Med Ekspert ; 67(3): 54-59, 2024.
Article in Russian | MEDLINE | ID: mdl-38887073

ABSTRACT

OBJECTIVE: To systematize the mechanisms of formation and morphology of secondary ischemic brain damage, formed in the case of craniocerebral injury. MATERIAL AND METHODS: A literature review devoted to the study of formation mechanisms of secondary ischemic brain damage in craniocerebral injury was conducted. The secondary sequential and secondary long-term ischemic brain damage in the posttraumatic period, as well as intracranial and extracranial factors that contribute to their occurrence and progression, were considered. RESULTS AND CONCLUSION: Analysis of the literature has shown that primary lesions occurring at the time of head injury must be differentiated from sequential and long-term secondary ischemic changes in different brain structures.


Subject(s)
Brain Ischemia , Craniocerebral Trauma , Humans , Craniocerebral Trauma/pathology , Craniocerebral Trauma/complications , Brain Ischemia/pathology , Brain Ischemia/etiology , Brain/pathology
6.
Chem Biol Interact ; 397: 111084, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38823537

ABSTRACT

Kaempferol is major flavonoid present in Convolvulus pluricaulis. This phytochemical protects the brain against oxidative stress, neuro-inflammation, neurotoxicity, neurodegeneration and cerebral ischemia induced neuronal destruction. Kaempferol is poorly water soluble. Our study proved that solid lipid nanoparticles (SLNs) were efficient carrier of kaempferol through blood-brain barrier (BBB). Kaempferol was incorporated into SLNs prepared from stearic acid with polysorbate 80 by the process of ultrasonication. Mean particle size and zeta potential of kaempferol loaded solid lipid nanoparticles (K-SLNs) were 451.2 nm and -15.0 mV. Atomic force microscopy showed that K-SLNs were spherical in shape. Fourier transformed infrared microscopy (FTIR) showed that both stearic acid and kaempferol were present in K-SLNs. X-ray diffraction (XRD) and differential scanning calorimetry (DSC) revealed that the matrices of K-SLNs were in untidy crystalline state. Entraptment efficiency of K-SLNs was 84.92%. In-vitro drug release percentage was 93.24%. Kaempferol loaded solid lipid nanoparticles (K-SLNs) showed controlled release profile. In-vitro uptake study showed significant efficiency of K-SLNs to cross blood-brain barrier (BBB). After oral administration into the focal cerebral ischemic rat, accumulation of fluorescent labeled K-SLNs was observed in the brain cortex which confirmed its penetrability into the brain. It significantly decreased the neurological deficit, infarct volume and level of reactive oxygen species (ROS) and decreased the level of pro-inflammatory mediators like NF-κB and p-STAT3. Damaged neurons and brain texture were improved. This study indicated increased bioavailability of kaempferol into the brain tissue through SLNs formulation.


Subject(s)
Blood-Brain Barrier , Brain Ischemia , Kaempferols , Nanoparticles , Animals , Kaempferols/chemistry , Kaempferols/administration & dosage , Kaempferols/pharmacology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Nanoparticles/chemistry , Rats , Male , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Brain Ischemia/pathology , Lipids/chemistry , Brain/metabolism , Brain/drug effects , Brain/pathology , Drug Carriers/chemistry , Particle Size , Rats, Wistar , Drug Liberation , NF-kappa B/metabolism , Liposomes
7.
Neurochem Res ; 49(8): 1965-1979, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38834843

ABSTRACT

Cerebral ischemia-reperfusion injury (CIRI) is the second leading cause of death worldwide, posing a huge risk to human life and health. Therefore, investigating the pathogenesis underlying CIRI and developing effective treatments are essential. Ferroptosis is an iron-dependent mode of cell death, which is caused by disorders in iron metabolism and lipid peroxidation. Previous studies demonstrated that ferroptosis is also a form of autophagic cell death, and nuclear receptor coactivator 4(NCOA4) mediated ferritinophagy was found to regulate ferroptosis by interfering with iron metabolism. Ferritinophagy and ferroptosis are important pathogenic mechanisms in CIRI. This review mainly summarizes the link and regulation between ferritinophagy and ferroptosis and further discusses their mechanisms in CIRI. In addition, the potential treatment methods targeting ferritinophagy and ferroptosis for CIRI are presented, providing new ideas for the prevention and treatment of clinical CIRI in the future.


Subject(s)
Ferritins , Ferroptosis , Reperfusion Injury , Ferroptosis/physiology , Humans , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Animals , Ferritins/metabolism , Iron/metabolism , Brain Ischemia/metabolism , Brain Ischemia/pathology , Nuclear Receptor Coactivators/metabolism , Autophagic Cell Death , Lipid Peroxidation/physiology
8.
Bull Exp Biol Med ; 176(6): 827-829, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38890211

ABSTRACT

The severity of ischemic injury was evaluated by densitometry of brain samples stained with 2,3,5-triphenyltetrazolium chloride (TTC) on a rat model of cerebral ischemia/reperfusion (common carotid artery occlusion) and the neuroprotective activity of an extract of Astragalus membranaceus, Scutellaria baicalensis, and Phlojodicarpus sibiricus was assessed. Occlusion of the common carotid arteries led to a weakening of TTC staining of the brain tissue: densitometric indicators of the staining intensity for the cortex and striatum were lower than the corresponding indicators of sham-operated rats by 18.3 and 10.4%. The mean intensity of staining of brain samples did not differ in rats treated with the extract and sham-operated animals, which attested to its neuroprotective effect. The applied method is convenient for evaluation of the severity of ischemic brain damage at the early stages and screening potential neuroprotective agents.


Subject(s)
Brain Ischemia , Neuroprotective Agents , Plant Extracts , Animals , Rats , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Male , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Astragalus propinquus/chemistry , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control , Tetrazolium Salts/chemistry , Brain/drug effects , Brain/pathology , Rats, Wistar , Disease Models, Animal , Scutellaria baicalensis
9.
Bull Exp Biol Med ; 176(6): 736-742, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38907060

ABSTRACT

Intranasal administration of total bovine brain gangliosides (6 mg/kg) to rats protected the CA1 hippocampal neurons from the death caused by two-vessel occlusion model (with hypotension) of forebrain ischemia/reperfusion injury. The immunohistochemical reaction of specific antibodies to marker proteins of activated microglia (Iba1) and astrocytes (GFAP) in hippocampal slices revealed the neuroprotective effect of exogenous gangliosides which can be mostly explained by their ability to suppress neuroinflammation and gliosis. The expression of neurotrophic factor BDNF in the CA1 region of hippocampus did not differ in sham-operated rats and animals exposed to ischemia/reperfusion. However, the administration of gangliosides increased the BDNF expression in both control and ischemic groups. The intranasal route of administration allows using lower concentrations of gangliosides preventing the death of hippocampal neurons.


Subject(s)
Administration, Intranasal , Brain-Derived Neurotrophic Factor , CA1 Region, Hippocampal , Gangliosides , Neurons , Neuroprotective Agents , Reperfusion Injury , Animals , Reperfusion Injury/pathology , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Gangliosides/pharmacology , Rats , Male , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/pathology , CA1 Region, Hippocampal/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/administration & dosage , Rats, Wistar , Glial Fibrillary Acidic Protein/metabolism , Calcium-Binding Proteins/metabolism , Microfilament Proteins/metabolism , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Brain Ischemia/metabolism , Prosencephalon/drug effects , Prosencephalon/pathology , Prosencephalon/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Cell Survival/drug effects , Disease Models, Animal
10.
J Cell Mol Med ; 28(12): e18449, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38924214

ABSTRACT

Mitochondrial dynamics has emerged as an important target for neuronal protection after cerebral ischaemia/reperfusion. Therefore, the aim of this study was to investigate the mechanism by which ARMC10 regulation of mitochondrial dynamics affects mitochondrial function involved in ischaemic stroke (IS). Mitochondrial morphology was detected by laser scanning confocal microscopy (LSCM), and mitochondrial ultrastructural alterations were detected by electron microscopy. The expression of mitochondrial dynamics-related genes Drp1, Mfn1, Mfn2, Fis1, OPA1 and ARMC10 and downstream target genes c-Myc, CyclinD1 and AXIN2 was detected by RT-qPCR. Western blot was used to detect the protein expression of ß-catenin, GSK-3ß, p-GSK-3ß, Bcl-2 and Bax. DCFH-DA fluorescent probe was to detect the effect of ARMC10 on mitochondrial ROS level, Annexin V-FITC fluorescent probe was to detect the effect of ARMC10 on apoptosis, and ATP assay kit was to detect the effect of ARMC10 on ATP production. Mitochondrial dynamics was dysregulated in clinical IS samples and in the OGD/R cell model, and the relative expression of ARMC10 gene was significantly decreased in IS group (p < 0.05). Knockdown and overexpression of ARMC10 could affect mitochondrial dynamics, mitochondrial function and neuronal apoptosis. Agonist and inhibitor affected mitochondrial function and neuronal apoptosis by targeting Wnt/ß-Catenin signal pathway. In the OGD/R model, ARMC10 affected mitochondrial function and neuronal apoptosis through the mechanism that regulates Wnt/ß-catenin signalling pathway. ARMC10 regulates mitochondrial dynamics and protects mitochondrial function by activating Wnt/ß-catenin signalling pathway, to exert neuroprotective effects.


Subject(s)
Apoptosis , Armadillo Domain Proteins , Ischemic Stroke , Mitochondria , Mitochondrial Dynamics , Wnt Signaling Pathway , Humans , Armadillo Domain Proteins/metabolism , Armadillo Domain Proteins/genetics , beta Catenin/metabolism , beta Catenin/genetics , Brain Ischemia/metabolism , Brain Ischemia/genetics , Brain Ischemia/pathology , Ischemic Stroke/metabolism , Ischemic Stroke/genetics , Ischemic Stroke/pathology , Mitochondria/metabolism , Reactive Oxygen Species/metabolism
11.
Eur J Pharmacol ; 977: 176718, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38849040

ABSTRACT

Nimodipine is used to prevent delayed ischemic deficit in patients with aneurysmal subarachnoid hemorrhage (aSAH). Spreading depolarization (SD) is recognized as a factor in the pathomechanism of aSAH and other acute brain injuries. Although nimodipine is primarily known as a cerebral vasodilator, it may have a more complex mechanism of action due to the expression of its target, the L-type voltage-gated calcium channels (LVGCCs) in various cells in neural tissue. This study was designed to investigate the direct effect of nimodipine on SD, ischemic tissue injury, and neuroinflammation. SD in control or nimodipine-treated live mouse brain slices was induced under physiological conditions using electrical stimulation, or by subjecting the slices to hypo-osmotic stress or mild oxygen-glucose deprivation (mOGD). SD was recorded applying local field potential recording or intrinsic optical signal imaging. Histological analysis was used to estimate tissue injury, the number of reactive astrocytes, and the degree of microglia activation. Nimodipine did not prevent SD occurrence in mOGD, but it did reduce the rate of SD propagation and the cortical area affected by SD. In contrast, nimodipine blocked SD occurrence in hypo-osmotic stress, but had no effect on SD propagation. Furthermore, nimodipine prevented ischemic injury associated with SD in mOGD. Nimodipine also exhibited anti-inflammatory effects in mOGD by reducing reactive astrogliosis and microglial activation. The results demonstrate that nimodipine directly inhibits SD, independent of nimodipine's vascular effects. Therefore, the use of nimodipine may be extended to treat acute brain injuries where SD plays a central role in injury progression.


Subject(s)
Brain Ischemia , Brain , Cortical Spreading Depression , Nimodipine , Animals , Nimodipine/pharmacology , Mice , Cortical Spreading Depression/drug effects , Male , Brain/drug effects , Brain/pathology , Brain/metabolism , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Mice, Inbred C57BL , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/pathology , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Calcium Channel Blockers/pharmacology , Calcium Channel Blockers/therapeutic use , Osmotic Pressure/drug effects
12.
PLoS One ; 19(6): e0305541, 2024.
Article in English | MEDLINE | ID: mdl-38885233

ABSTRACT

BACKGROUND: The inflammatory response is a key factor in the pathogenesis of cerebral ischemia/reperfusion injury (CIRI), and anti-inflammatory interventions may offer a promising therapeutic strategy. Forsythoside B (FB) is a phenylethanoid glycoside isolated from Forsythiae fructus, which has been reported to have anti-inflammatory effects. However, the mechanism of the neuroprotective effect of FB on CIRI remains unclear. METHODS: Adult male Sprague-Dawley rats were subjected to transient middle cerebral artery occlusion/reperfusion (MCAO/R). FB was administered intraperitoneally for 3 days prior to MCAO/R. Cerebral infarct volume and neurological deficit score were used as indices to evaluate MCAO/R injury. The serum levels of inflammatory factors and antioxidant enzymes were measured. The activation of silent information regulator 2 homolog 1 (Sirt1) and the inhibition of the nucleotide-binding oligomerization domain-like receptor with a pyrin domain 3 (NLRP3) pathway were assessed through western blot and immunohistochemistry analysis. Furthermore, the rats were treated with Sirt1 shRNA 3 days before MCAO/R by stereotactical injection into the ipsilateral hemispheric region to assess the impact of Sirt1 knockdown on the protection of FB during MCAO/R. RESULTS: FB reduced cerebral infarct volume and neurological deficit score in MCAO/R rats. FB reduced pathological changes and cell apoptosis in the hippocampal CA1 region and cortex on the ischemic side of rats. FB inhibited the serum levels of inflammatory factors and increased the activities of antioxidant enzymes. Further study showed that FB inhibited the activation of the NLRP3 pathway and induced Sirt1 activation. CONCLUSION: FB demonstrated neuroprotective and anti-inflammatory effects by inhibiting the NLRP3 pathway through Sirt1 activation in CIRI.


Subject(s)
Infarction, Middle Cerebral Artery , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Rats, Sprague-Dawley , Reperfusion Injury , Sirtuin 1 , Animals , Sirtuin 1/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Male , Inflammasomes/metabolism , Rats , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/complications , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Brain Ischemia/pathology , Caffeic Acids , Glucosides
13.
Eur J Med Res ; 29(1): 347, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38926868

ABSTRACT

OBJECTIVE: α-HBDH serves as a biomarker of myocardial damage and is implicated in adverse outcomes across various critical illnesses. Our study aimed to assess the correlation between α-HBDH levels, and severity and recurrence of acute ischemic stroke (AIS). METHODS: We enrolled patients with mild-to-moderate AIS within 72 h of onset. Based on the baseline score of the National Institutes of Health Stroke Scale (bNIHSS) at registration, patients were categorized into mild (bNIHSS ≤ 4 points) and moderate AIS groups (4 < bNIHSS ≤ 10 points). Subsequently, based on the normal upper limit of α-HBDH, patients were divided into low-level α-HBDH (≤ 180 U/L) and high-level α-HBDH (> 180 U/L) groups. Multivariate logistic regression analysis and Cox proportional hazard regression analysis were employed to evaluate the relationship between α-HBDH levels and bNIHSS scores as well as the risk of recurrent AIS within 90 days. RESULTS: We observed a significant association between higher baseline levels of α-HBDH and increased bNIHSS scores, indicating a more severe AIS (odds ratio = 24.449; 95% confidence interval [CI], 8.749-68.324; p < 0.01). Additionally, the risk of recurrent AIS within 90 days was 4.666 times higher in the high-level α-HBDH group compared to the low-level group (hazard ratio = 4.666; 95% CI, 2.481-8.777; p < 0.01). CONCLUSIONS: The baseline level of α-HBDH is significantly correlated with the severity of AIS and the risk of recurrent AIS within 90 days.


Subject(s)
Ischemic Stroke , Recurrence , Severity of Illness Index , Humans , Male , Female , Ischemic Stroke/diagnosis , Ischemic Stroke/etiology , Middle Aged , Aged , Biomarkers/blood , Brain Ischemia/etiology , Brain Ischemia/pathology
14.
Bull Exp Biol Med ; 176(5): 649-657, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38733482

ABSTRACT

In translational animal study aimed at evaluation of the effectiveness of innovative methods for treating cerebral stroke, including regenerative cell technologies, of particular importance is evaluation of the dynamics of changes in the volume of the cerebral infarction in response to therapy. Among the methods for assessing the focus of infarction, MRI is the most effective and convenient tool for use in preclinical studies. This review provides a description of MR pulse sequences used to visualize cerebral ischemia at various stages of its development, and a detailed description of the MR semiotics of cerebral infarction. A comparison of various methods for morphometric analysis of the focus of a cerebral infarction, including systems based on artificial intelligence for a more objective measurement of the volume of the lesion, is also presented.


Subject(s)
Magnetic Resonance Imaging , Magnetic Resonance Imaging/methods , Animals , Stroke/diagnostic imaging , Stroke/pathology , Brain Ischemia/diagnostic imaging , Brain Ischemia/pathology , Disease Models, Animal , Cerebral Infarction/diagnostic imaging , Cerebral Infarction/pathology , Ischemic Stroke/diagnostic imaging , Ischemic Stroke/pathology , Artificial Intelligence
15.
Cell Biol Toxicol ; 40(1): 31, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38767771

ABSTRACT

Mitochondrial dysfunction contributes to cerebral ischemia-reperfusion (CI/R) injury, which can be ameliorated by Sirtuin-3 (SIRT3). Under stress conditions, the SIRT3-promoted mitochondrial functional recovery depends on both its activity and expression. However, the approach to enhance SIRT3 activity after CI/R injury remains unelucidated. In this study, Sprague-Dawley (SD) rats were intracranially injected with either adeno-associated viral Sirtuin-1 (AAV-SIRT1) or AAV-sh_SIRT1 before undergoing transient middle cerebral artery occlusion (tMCAO). Primary cortical neurons were cultured and transfected with lentiviral SIRT1 (LV-SIRT1) and LV-sh_SIRT1 respectively before oxygen-glucose deprivation/reoxygenation (OGD/R). Afterwards, rats and neurons were respectively treated with a selective SIRT3 inhibitor, 3-(1H-1,2,3-triazol-4-yl) pyridine (3-TYP). The expression, function, and related mechanism of SIRT1 were investigated by Western Blot, flow cytometry, immunofluorescence staining, etc. After CI/R injury, SIRT1 expression decreased in vivo and in vitro. The simulation and immune-analyses reported strong interaction between SIRT1 and SIRT3 in the cerebral mitochondria before and after CI/R. SIRT1 overexpression enhanced SIRT3 activity by increasing the deacetylation of SIRT3, which ameliorated CI/R-induced cerebral infarction, neuronal apoptosis, oxidative stress, neurological and motor dysfunction, and mitochondrial respiratory chain dysfunction, promoted mitochondrial biogenesis, and retained mitochondrial integrity and mitochondrial morphology. Meanwhile, SIRT1 overexpression alleviated OGD/R-induced neuronal death and mitochondrial bioenergetic deficits. These effects were reversed by AAV-sh_SIRT1 and the neuroprotective effects of SIRT1 were partially offset by 3-TYP. These results suggest that SIRT1 restores the structure and function of mitochondria by activating SIRT3, offering neuroprotection against CI/R injury, which signifies a potential approach for the clinical management of cerebral ischemia.


Subject(s)
Brain Ischemia , Mitochondria , Neurons , Rats, Sprague-Dawley , Reperfusion Injury , Sirtuin 1 , Sirtuin 3 , Animals , Sirtuin 1/metabolism , Sirtuin 1/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Mitochondria/metabolism , Male , Sirtuin 3/metabolism , Sirtuin 3/genetics , Neurons/metabolism , Neurons/pathology , Rats , Brain Ischemia/metabolism , Brain Ischemia/pathology , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Apoptosis , Sirtuins
16.
Int J Med Sci ; 21(7): 1274-1279, 2024.
Article in English | MEDLINE | ID: mdl-38818467

ABSTRACT

Objective: Citicoline can be used to reduce acute ischemic stroke injury via venous infusion, however, its protective effects in the brain extracellular space remain largely unknown. Herein, we investigated the brain protective effects of citicoline administered via the brain extracellular space and sought precise effective dosage range that can protect against ischemic injury after experimental ischemic stroke in rats. Methods: Fifty-six Sprague-Dawley rats were randomly divided into control, intraperitoneal (IP), caudate-putamen (CPu)-25, CPu-40, CPu-50, CPu-60 and CPu-75 groups based on the infusion site and concentration of citicoline. Two hours after the administration of citicoline, the rats were subjected to a permanent middle cerebral artery occlusion to mimic acute ischemic stroke. Then, the brain infarct volume in rats after stroke was measured and their neurological deficiency was evaluated to explain the protective effects and effective dosage range of citicoline. Results: Compared to the control and IP groups, brain infarct volume of rats in CPu-40, CPu-50, and CPu-60 groups is significant smaller. Furthermore, the brain infarct volume of rats in CPu-50 is the least. Conclusions: Here, we showed that citicoline can decrease the brain infarct volume, thus protecting the brain from acute ischemic stroke injury. We also found that the appropriate effective citicoline dose delivered via the brain extracellular space is 50 mM. Our study provides novel insights into the precise treatment of acute ischemic stroke by citicoline via the brain extracellular space, further guiding the treatment of brain disease.


Subject(s)
Brain , Cytidine Diphosphate Choline , Disease Models, Animal , Extracellular Space , Ischemic Stroke , Rats, Sprague-Dawley , Animals , Cytidine Diphosphate Choline/administration & dosage , Cytidine Diphosphate Choline/pharmacology , Cytidine Diphosphate Choline/therapeutic use , Rats , Ischemic Stroke/drug therapy , Ischemic Stroke/pathology , Extracellular Space/drug effects , Male , Brain/drug effects , Brain/pathology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Humans , Infarction, Middle Cerebral Artery/drug therapy , Brain Ischemia/drug therapy , Brain Ischemia/pathology
17.
Cell Mol Life Sci ; 81(1): 224, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38769196

ABSTRACT

Synaptic loss is an early event in the penumbra area after an ischemic stroke. Promoting synaptic preservation in this area would likely improve functional neurological recovery. We aimed to detect proteins involved in endogenous protection mechanisms of synapses in the penumbra after stroke and to analyse potential beneficial effects of these candidates for a prospective stroke treatment. For this, we performed Liquid Chromatography coupled to Mass Spectrometry (LC-MS)-based proteomics of synaptosomes isolated from the ipsilateral hemispheres of mice subjected to experimental stroke at different time points (24 h, 4 and 7 days) and compared them to sham-operated mice. Proteomic analyses indicated that, among the differentially expressed proteins between the two groups, cystatin C (CysC) was significantly increased at 24 h and 4 days following stroke, before returning to steady-state levels at 7 days, thus indicating a potential transient and intrinsic rescue mechanism attempt of neurons. When CysC was applied to primary neuronal cultures subjected to an in vitro model of ischemic damage, this treatment significantly improved the preservation of synaptic structures. Notably, similar effects were observed when CysC was loaded into brain-derived extracellular vesicles (BDEVs). Finally, when CysC contained in BDEVs was administered intracerebroventricularly to stroked mice, it significantly increased the expression of synaptic markers such as SNAP25, Homer-1, and NCAM in the penumbra area compared to the group supplied with empty BDEVs. Thus, we show that CysC-loaded BDEVs promote synaptic protection after ischemic damage in vitro and in vivo, opening the possibility of a therapeutic use in stroke patients.


Subject(s)
Brain Ischemia , Brain , Cystatin C , Extracellular Vesicles , Mice, Inbred C57BL , Synapses , Animals , Extracellular Vesicles/metabolism , Extracellular Vesicles/transplantation , Cystatin C/metabolism , Synapses/metabolism , Mice , Male , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain/metabolism , Brain/pathology , Proteomics/methods , Synaptosomes/metabolism , Neurons/metabolism , Stroke/metabolism , Stroke/pathology , Stroke/therapy , Cells, Cultured , Disease Models, Animal
18.
Neurochem Res ; 49(7): 1863-1878, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38753259

ABSTRACT

The study aimed to assess 𝛾-Terpinene's (𝛾-TER) neuroprotective potential in acute cerebral ischemia, characterized by reduced cerebral blood flow in rats. Middle cerebral artery occlusion (MCAO), a standard method for inducing cerebral ischemia, was employed in male Wistar rats. 𝛾-TER at varying doses (5, 10, and 15 mg/kg) were intraperitoneally administered during reperfusion onset. Neurological outcomes, cerebral infarct size, edema, and enzymatic activities (SOD, GPx, and catalase) in the brain were evaluated using diverse techniques. The study examined gene expression and pathways associated with neuroinflammation and apoptosis using Cytoscape software, identifying the top 10 genes involved. Pro-inflammatory and pro-apoptotic factors were assessed through real-time PCR and ELISA, while apoptotic cell rates were measured using the TUNEL and Flow cytometry assay. Immunohistochemistry assessed apoptosis-related proteins like Bax and bcl-2 in the ischemic area. 𝛾-TER, particularly at doses of 10 and 15 mg/kg, significantly reduced neurological deficits and cerebral infarction size. The 15 mg/kg dose mitigated TNF-α, IL-1ß, Bax, and caspase-3 gene and protein levels in the cortex, hippocampus, and striatum compared to controls. Furthermore, Bcl-2 levels increased in these regions. 𝛾-TER show cased neuroprotective effects by suppressing inflammation, apoptosis, and oxidation. In conclusion, 𝛾-TER, possessing natural anti-inflammatory and anti-apoptotic properties, shields the brain against ischemic damage by reducing infarction, edema, oxidative stress, and inflammation. It modulates the expression of crucial genes and proteins associated with apoptosis in diverse brain regions. These findings position 𝛾-TER as a potential therapeutic agent for ischemic stroke.


Subject(s)
Apoptosis , Neuroprotective Agents , Rats, Wistar , Animals , Male , Apoptosis/drug effects , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Rats , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Brain Ischemia/pathology , Oxidative Stress/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Cyclohexane Monoterpenes/therapeutic use , Cyclohexane Monoterpenes/pharmacology , Oxidation-Reduction/drug effects , Brain/drug effects , Brain/metabolism , Brain/pathology
19.
Stroke ; 55(6): 1650-1659, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38738428

ABSTRACT

BACKGROUND: Beyond neuronal injury, cell death pathways may also contribute to vascular injury after stroke. We examined protein networks linked to major cell death pathways and identified SLC22A17 (solute carrier family 22 member 17) as a novel mediator that regulates endothelial tight junctions after ischemia and inflammatory stress. METHODS: Protein-protein interactions and brain enrichment analyses were performed using STRING, Cytoscape, and a human tissue-specific expression RNA-seq database. In vivo experiments were performed using mouse models of transient focal cerebral ischemia. Human stroke brain tissues were used to detect SLC22A17 by immunostaining. In vitro experiments were performed using human brain endothelial cultures subjected to inflammatory stress. Immunostaining and Western blot were used to assess responses in SLC22A17 and endothelial tight junctional proteins. Water content, dextran permeability, and electrical resistance assays were used to assess edema and blood-brain barrier (BBB) integrity. Gain and loss-of-function studies were performed using lentiviral overexpression of SLC22A17 or short interfering RNA against SLC22A17, respectively. RESULTS: Protein-protein interaction analysis showed that core proteins from apoptosis, necroptosis, ferroptosis, and autophagy cell death pathways were closely linked. Among the 20 proteins identified in the network, the iron-handling solute carrier SLC22A17 emerged as the mediator enriched in the brain. After cerebral ischemia in vivo, endothelial expression of SLC22A17 increases in both human and mouse brains along with BBB leakage. In human brain endothelial cultures, short interfering RNA against SLC22A17 prevents TNF-α (tumor necrosis factor alpha)-induced ferroptosis and downregulation in tight junction proteins and disruption in transcellular permeability. Notably, SLC22A17 could repress the transcription of tight junctional genes. Finally, short interfering RNA against SLC22A17 ameliorates BBB leakage in a mouse model of focal cerebral ischemia. CONCLUSIONS: Using a combination of cell culture, human stroke samples, and mouse models, our data suggest that SLC22A17 may play a role in the control of BBB function after cerebral ischemia. These findings may offer a novel mechanism and target for ameliorating BBB injury and edema after stroke.


Subject(s)
Blood-Brain Barrier , Brain Ischemia , Tight Junctions , Aged , Animals , Female , Humans , Male , Mice , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain Ischemia/genetics , Cell Death , Endothelial Cells/metabolism , Mice, Inbred C57BL , Organic Cation Transport Proteins/metabolism , Organic Cation Transport Proteins/genetics , Tight Junctions/metabolism
20.
Cell Rep Med ; 5(5): 101522, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38701781

ABSTRACT

Neuroinflammation plays a significant role in ischemic injury, which can be promoted by oxidized mitochondrial DNA (Ox-mtDNA). Cytidine/uridine monophosphate kinase 2 (CMPK2) regulates mtDNA replication, but its role in neuroinflammation and ischemic injury remains unknown. Here, we report that CMPK2 expression is upregulated in monocytes/macrophages and microglia post-stroke in humans and mice, respectively. Microglia/macrophage CMPK2 knockdown using the Cre recombination-dependent adeno-associated virus suppresses the inflammatory responses in the brain, reduces infarcts, and improves neurological outcomes in ischemic CX3CR1Cre/ERT2 mice. Mechanistically, CMPK2 knockdown limits newly synthesized mtDNA and Ox-mtDNA formation and subsequently blocks NLRP3 inflammasome activation in microglia/macrophages. Nordihydroguaiaretic acid (NDGA), as a CMPK2 inhibitor, is discovered to reduce neuroinflammation and ischemic injury in mice and prevent the inflammatory responses in primary human monocytes from ischemic patients. Thus, these findings identify CMPK2 as a promising therapeutic target for ischemic stroke and other brain disorders associated with neuroinflammation.


Subject(s)
Ischemic Stroke , Microglia , Neuroinflammatory Diseases , Animals , Humans , Male , Mice , Brain Injuries/pathology , Brain Injuries/metabolism , Brain Injuries/genetics , Brain Ischemia/pathology , Brain Ischemia/metabolism , Brain Ischemia/genetics , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Inflammasomes/metabolism , Ischemic Stroke/pathology , Ischemic Stroke/metabolism , Ischemic Stroke/genetics , Macrophages/metabolism , Macrophages/pathology , Mice, Inbred C57BL , Microglia/metabolism , Microglia/pathology , Monocytes/metabolism , Monocytes/drug effects , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...