Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Sci Rep ; 14(1): 5580, 2024 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-38448479

RESUMO

Inflammation and oxidative stress are becoming more recognized as risk factors for ischemic stroke. Paeoniflorigenone (PA) has diverse pharmacological effects that include anti-inflammatory and antioxidant properties. However, the specific mechanisms by which PA affects cerebral ischemic stroke have not been studied. Our objective was to investigate the potential targets and mechanisms of PA in preventing cerebral ischemic stroke. We obtained the potential targets of PA from the SwissTargetPrediction, Super-PRED, and SEA Search Server databases. The GSE97537 dataset was utilized to identify gene targets related to ischemic stroke. The overlapping targets were imported into the STRING database to construct a protein-protein interaction network, and enrichment analyses were conducted using R software. Rats were pretreated with PA for three weeks before undergoing MCAO and reperfusion. H&E staining, ELISA, and qRT-PCR analyses were then performed to explore the potential mechanisms of PA. In the study, we identified 439 potential targets for PA and 1206 potential targets for ischemic stroke. Out of these, there were 71 common targets, which were found to be primarily associated with pathways related to oxidative stress and inflammation. The results from animal experiments showed that PA was able to improve nerve function and reduce inflammatory cytokines and oxidative stress in the MCAO-induced ischemic stroke model. Additionally, the expression of core genes in the MCAO + HPA group was significantly lower compared to the MCAO group. Our study revealed that the potential mechanisms by which PA prevents ischemic stroke involve oxidative stress and inflammation. These findings provide important theoretical guidance for the clinical use of PA in preventing and managing ischemic stroke.


Assuntos
AVC Isquêmico , Monoterpenos , Acidente Vascular Cerebral , Animais , Ratos , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/prevenção & controle , Inflamação/tratamento farmacológico , Estresse Oxidativo , Biologia Computacional
2.
Environ Toxicol ; 39(3): 1531-1543, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38009636

RESUMO

Cerebral ischemia and reperfusion are related to various situations like injuries after various traumas, oxidative stress, increased calcium ion, capillary hypoperfusion, microvascular hyperpermeability, leukocyte infiltration, and blood-brain barrier disruption. An antidepressant Agomelatine which is a melatonin receptor (MT1/MT2) agonist and serotonin receptor (5-HT2C) antagonist has been reported by studies to have antioxidant and anti-inflammatory effects. In our study, we aimed to detect the effects of citrate-coated silver nanoparticle-loaded agomelatine application on neurodegeneration, endoplasmic reticulum stress, autophagic and apoptotic cell death, inflammation, and P2X7R expression in the cerebral ischemia-reperfusion model to facilitate the passage of blood-brain barrier. Forty two Sprague-Dawley rats in total were divided into six equal groups (n:7) and applications were performed. Acute cerebral injury in the ischemia-reperfusion model was created 2 h after internal carotid artery ligation in rats and then at the 2nd hour of reperfusion citrate-coated silver nanoparticles loaded with Agomelatine were applied. Twenty four hours later, neurologic analysis on animals in experimental groups was performed, animals were decapitated and GSH, GPx, SOD, CAT, MDA, IL-1ß, and TNF-α parameters were examined after taking blood and the cerebral tissue samples. As a result, it was determined that ischemia-reperfusion caused endoplasmic reticulum stress in the cerebral tissues and thus caused cellular injury.


Assuntos
Isquemia Encefálica , Nanopartículas Metálicas , Traumatismo por Reperfusão , Ratos , Animais , Ratos Sprague-Dawley , Inflamassomos/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Prata , Ácido Cítrico/farmacologia , Traumatismo por Reperfusão/metabolismo , Estresse Oxidativo , Isquemia Encefálica/metabolismo , Citratos/farmacologia , Reperfusão , Isquemia , Estresse do Retículo Endoplasmático
3.
Chinese Pharmacological Bulletin ; (12): 263-272, 2024.
Artigo em Chinês | WPRIM (Pacífico Ocidental) | ID: wpr-1013625

RESUMO

Aim To investigate the dynamic time-course changes in neuronal cytoskeleton after acute ischemia and reperfusion in rats. Methods Reperfusion was performedin rats by blocking the middle cerebralarteryfor 90 min, then therats wereobserved and collected at different time points. The brain damage wasobserved by Nissl staining,and neurobehavioural function was evaluated with neurological deficit score and forelimb placement test. The cellular changes in the alternations of cytoskeletal elements including microtubule associated protein 2 (MAP2) and neurofilament heavy chain (NF-H) were observed by immunohistochemistry staining and Western blot. Impaired axons, dendrites and cytoskeletal alternations were detected by electron microscope. Results Brain damage and neurobehavioural function were gradually aggravated with the prolongation of reperfusion. Brain damage appeared earlier and more severe in striatum than in cortex. Moreover, decreased MAP2-related and increased NF-H-related immunoreactive intensities were found in the ischemic areas. Impaired cytoskeletal arrangement and reduced dense were indicated. Damaged cytoskeletal components such as microtubules and neurofilament arrangement, decreased axonal filament density, and swelled dendrites were observed after cerebral ischemia reperfusion by ultrastructural observations. Conclusions Different brain regions have diverse tolerance to ischemia-reperfusion injury. Major elements of neuronal cytoskeleton show dynamic responses to ischemia and reperfusion, which may further contribute to brain damage and neurological impairment following MCAO and reperfusion.

4.
Biomed Pharmacother ; 167: 115492, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37716121

RESUMO

Cardiopulmonary resuscitation and related life support technologies have improved substantially in recent years; however, mortality and disability rates from cardiac arrest (CA) remain high and are closely associated with the high incidence of cerebral ischemia-reperfusion injury (CIRI), which is explained by a "double-hit" model (i.e., resulting from both ischemia and reperfusion). Mitochondria are important power plants in the cell and participate in various biochemical processes, such as cell differentiation and signaling in eukaryotes. Various mitochondrial processes, including energy metabolism, calcium homeostasis, free radical production, and apoptosis, are involved in several important stages of the progression and development of CIRI. Mitophagy is a key mechanism of the endogenous removal of damaged mitochondria to maintain organelle function and is a critical target for CIRI treatment after CA. Mitophagy also plays an essential role in attenuating ischemia-reperfusion in other organs, particularly during post-cardiac arrest myocardial dysfunction. Regulation of mitophagy may influence necroptosis (a programmed cell death pathway), which is the main endpoint of organ ischemia-reperfusion injury. In this review, we summarize the main signaling pathways related to mitophagy and their associated regulatory proteins. New therapeutic methods and drugs targeting mitophagy in ischemia-reperfusion animal models are also discussed. In-depth studies of the mechanisms underlying the regulation of mitophagy will enhance our understanding of the damage and repair processes in CIRI after CA, thereby contributing to the development of new therapeutic strategies.


Assuntos
Parada Cardíaca , Traumatismo por Reperfusão , Ratos , Animais , Mitofagia , Ratos Sprague-Dawley , Traumatismo por Reperfusão/metabolismo , Isquemia , Parada Cardíaca/complicações
5.
Cell Signal ; 109: 110751, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37321527

RESUMO

Microglia-mediated inflammation is a major contributor to the brain damage in cerebral ischemia and reperfusion (I/R) injury, and N6-Methyladenosine (m6A) has been implicated in cerebral I/R injury. Here, we explored whether m6A modification is associated with microglia-mediated inflammation in cerebral I/R injury and its underlying regulatory mechanism using an in vivo mice model of intraluminal middle cerebral artery occlusion/reperfusion (MCAO/R) and in vitro models of primary isolated microglia and BV2 microglial cells subjected to oxygen-glucose deprivation and reoxygenation (OGD/R) were used. We found microglial m6A modification increased and microglial fat mass and obesity-associated protein (FTO) expression decreased in cerebral I/R injury in vivo and in vitro. Inhibition of m6A modification by intraperitoneal injection of Cycloleucine (Cyc) in vivo or transfection of FTO plasmid in vitro significantly alleviated brain injury and microglia-mediated inflammatory response. Through Methylated RNA immunoprecipitation sequencing (MeRIP-Seq), RNA sequencing (RNA-Seq) and western blotting, we found that m6A modification promoted cerebral I/R-induced microglial inflammation via increasing cGAS mRNA stability to aggravate Sting/NF-κB signaling. In conclusion, this study deepens our understanding on the relationship of m6A modification and microglia-mediated inflammation in cerebral I/R injury, and insights a novel m6A-based therapeutic for inhibiting inflammatory response against ischemic stroke.


Assuntos
Isquemia Encefálica , Traumatismo por Reperfusão , Camundongos , Animais , Doenças Neuroinflamatórias , Isquemia Encefálica/metabolismo , Transdução de Sinais/fisiologia , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/metabolismo , Microglia/metabolismo , Inflamação/metabolismo , Reperfusão , Dioxigenase FTO Dependente de alfa-Cetoglutarato
6.
ACS Chem Neurosci ; 14(11): 2146-2158, 2023 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-37170554

RESUMO

Cerebral ischemia/reperfusion (I/R) injury is a key reason for the poor prognosis of ischemic stroke. As only a few neuroprotective medications for cerebral I/R injury have been applied in the clinic, it is necessary to design a new therapeutic strategy to treat cerebral I/R injury. The N-salicyloyl tryptamine derivative LZWL02003, synthesized from melatonin and salicylic acid, exhibits a wide range of biological properties. In this study, we assessed the neuroprotective capabilities of LZWL02003 in vivo and in vitro and investigated its possible mechanisms. Oxygen-glucose deprivation/reoxygenation was utilized to create an in vitro model of cerebral I/R damage. Middle cerebral artery occlusion/reperfusion was employed to imitate cerebral I/R injury in vivo. Neuronal apoptosis, oxidative stress, mitochondrial dysfunction, and neuroinflammation are associated with the pathogenesis of cerebral I/R injury. Our findings demonstrated that LZWL02003 upregulated the expression of Bcl-2 and downregulated the expression of Bax, thus maintaining the homeostasis of Bcl-2/Bax proteins and preventing apoptosis. LZWL02003 also reduced oxidative stress by reducing malondialdehyde and reactive oxygen species levels, increasing the superoxide dismutase activity, and resolving mitochondrial malfunction. LZWL02003 can lower interleukin (IL)-1ß, tumor necrosis factor (TNF)-α, and IL-6 levels, which in turn suppress neuroinflammation. Activation of the nuclear factor-kappa B (NF-κB) pathway is involved in various pathophysiologies, including cerebral I/R injury. We discovered that LZWL02003 suppressed the phosphorylation activation of NF-κB pathway-related proteins and decreased the nuclear translocation of NF-κB p65 subunits. Taken together, our results suggest that LZWL02003 is a neuroprotective drug with pleiotropic effects and may be a candidate for treating cerebral I/R injury.


Assuntos
Isquemia Encefálica , Fármacos Neuroprotetores , Traumatismo por Reperfusão , Humanos , NF-kappa B/metabolismo , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Proteína X Associada a bcl-2 , Doenças Neuroinflamatórias , Traumatismo por Reperfusão/metabolismo , Isquemia Encefálica/tratamento farmacológico , Infarto da Artéria Cerebral Média/tratamento farmacológico , Proteínas Proto-Oncogênicas c-bcl-2 , Triptaminas/farmacologia , Apoptose
7.
Pharmaceuticals (Basel) ; 16(4)2023 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-37111367

RESUMO

Neuroprotective drugs to protect the brain against cerebral ischemia and reperfusion (I/R) injury are urgently needed. Mammalian cell-produced recombinant human erythropoietin (rhuEPOM) has been demonstrated to have excellent neuroprotective functions in preclinical studies, but its neuroprotective properties could not be consistently translated in clinical trials. The clinical failure of rhuEPOM was thought to be mainly due to its erythropoietic activity-associated side effects. To exploit its tissue-protective property, various EPO derivatives with tissue-protective function only have been developed. Among them, asialo-rhuEPO, lacking terminal sialic acid residues, was shown to be neuroprotective but non-erythropoietic. Asialo-rhuEPO can be prepared by enzymatic removal of sialic acid residues from rhuEPOM (asialo-rhuEPOE) or by expressing human EPO gene in glycoengineered transgenic plants (asialo-rhuEPOP). Both types of asialo-rhuEPO, like rhuEPOM, displayed excellent neuroprotective effects by regulating multiple cellular pathways in cerebral I/R animal models. In this review, we describe the structure and properties of EPO and asialo-rhuEPO, summarize the progress on neuroprotective studies of asialo-rhuEPO and rhuEPOM, discuss potential reasons for the clinical failure of rhuEPOM with acute ischemic stroke patients, and advocate future studies needed to develop asialo-rhuEPO as a multimodal neuroprotectant for ischemic stroke treatment.

8.
Chinese Pharmacological Bulletin ; (12): 739-744, 2023.
Artigo em Chinês | WPRIM (Pacífico Ocidental) | ID: wpr-1013940

RESUMO

Aim To observe cellular damage and astrocyte activation at different time points of cerebral ischemia and reperfusion. Methods The middle cerebral artery of male SpragueDawley rats was occluded for 90 min followed by different time points of reperfusion. Eighty-five SPF male SD rats were randomly divided into control group (Sham), IR3, 6, 12, 24 and IR48h (MCAO followed by 48 h of reperfusion) group. Cerebral ischemia and reperfusion injury was observed by HE staining, and the structure of astrocytes was estimated with transmission electron microscopy (TEM). GFAP expression was detected by immunofluorescence staining and Western blot. Results Cerebral ischemia following by different time points of reperfusion led to different degrees of cellular damage, which was the most serious at 24 h of reperfusion. TEM showed destruction of astrocytes structure, swollen organelles and broken mitochondrial ridge. After cerebral ischemia-reperfusion, the expression levels of GFAP were significant up-regulated in the ischemic penumbra cortex and the highest was at 48 h of reperfusion, indicating astrocytes were activated. In addition, the results showed the gradual decrease in GFAP expression in the infarct core. Conclusions After cerebral ischemia-reperfusion, cellular damage is aggravated, and astrocytes are gradually activated in the ischemic penumbra. With the extension of reperfusion time, the boundaries of infarct area and ischemic area are gradually clear, and scarring may occur.

9.
Neurochem Res ; 47(5): 1369-1382, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35099719

RESUMO

Hyperglycemia aggravates cerebral ischemia/reperfusion (I/R) injury via vascular injury. There is still a lack of effective pharmaceutical preparations for cerebral I/R injury under hyperglycemia. This study aimed to investigate the effects of oxymatrine (OMT) on hyperglycemia-exacerbated cerebral I/R injury in vitro and in vivo. The middle cerebral artery occlusion (MCAO) and reperfusion was established in the rats under hyperglycemia. Meanwhile, oxygen-glucose deprivation and reoxygenation (OGD/R) with high glucose was used as an in vitro model of hyperglycemic cerebral I/R injury. The results showed that the neurological deficit score, mortality, infarct volume and penumbra apoptosis in hyperglycemia group were significantly higher than those in normal glucose group. OMT pre-treated obviously reduced the degree of neurological deficit, mortality, infarct volume, improve cerebral blood flow after I/R in rats with hyperglycemia, and increase the survival rate of human brain microvascular endothelial cells (HBMECs) in high glucose and OGD/R group. OMT significantly improved the ultrastructure changes of endothelial cells, and maintain the migration and angiogenesis potency of HBMECs in high glucose and OGD/R group. OMT obviously alleviated the down-regulating CD31 and CD105 expression in cerebral microvessels caused by hyperglycemia. It is concluded that OMT treatment might alleviate cerebral I/R injury under hyperglycemia via protecting microvessels.


Assuntos
Alcaloides , Isquemia Encefálica , Quinolizinas , Traumatismo por Reperfusão , Alcaloides/uso terapêutico , Animais , Apoptose , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/metabolismo , Células Endoteliais/metabolismo , Humanos , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/metabolismo , Microvasos/metabolismo , Quinolizinas/uso terapêutico , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/metabolismo
10.
Artigo em Chinês | WPRIM (Pacífico Ocidental) | ID: wpr-933946

RESUMO

Objective:To explore the effect of hyperbaric oxygen (HBO) on the blood-brain barrier via the silent information regulator 1 (SIRT1)/Forkhead box O1(FoxO1) signaling pathway after cerebral ischemia and reperfusion using a rat model.Methods:Forty Wistar rats were randomly assigned into sham, cerebral ischemia-reperfusion (CIR), CIR+ HBO and CIR+ HBO+ EX527 groups, each of 10. The cerebral ischemia-reperfusion model was established in all groups except the sham group by right middle cerebral artery occlusion using the modified thread-occlusion method. The sham group was not ligated. Both the CIR+ HBO and CIR+ HBO+ EX527 groups were given HBO 1, 9, 21, 45 and 69 hours after the reperfusion. The CIR+ HBO+ EX527 group was additionally injected with 5mg/kg of EX527(a SIRT1inhibitor) peritoneally 4, 12, 24, 48 and 72 hours after the reperfusion. Then 2% Evens blue (EB) was injected into the tail vein an hour before the rats were sacrificed. The content of EB and the expression of SIRT1, FoxO1, ZO-1, Occludin, Claudin-5 mRNA and their proteins were determined using spectrophotometry, reverse transcription-polymerase chain reactions and Western blotting.Results:The average EB content of the hippocampal brain tissue from the CIR, CIR+ HBO and CIR+ HBO+ EX527 rats was significantly greater than the Sham group′s average 72h after reperfusion. The average expression of SIRT1, FoxO1, ZO-1, Occludin and Claudin-5 mRNA and their proteins was significantly lower, with the CIR + HBO + EX 527 group′s average significantly lower than that of the CIR+ HBO group.Conclusions:HBO can increase the expression of tight junction protein via the SIRT1/FoxO1 pathway. It helps to protect the blood-brain barrier in CIR injury situations.

11.
Int J Mol Sci ; 22(18)2021 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-34575823

RESUMO

To investigate the effect of 14,15-EET on the parthanatos in neurons induced by cerebral ischemia and reperfusion, middle cerebral artery occlusion and reperfusion (MCAO/R) and oxygen glucose deprivation/reoxygenation (OGD/R) were used to simulate cerebral ischemia reperfusion in vivo and in vitro, respectively. TTC staining and the Tunel method were used to detect cerebral infarct volume and neuronal apoptosis. Western blot and immunofluorescence were used to detect poly (ADP-ribose) polymerase-1 (PARP-1) activation and AIF nuclear translocation. The production of reactive oxygen species (ROS) and the expression of antioxidant genes were detected by Mito SOX, DCFH-DA and qPCR methods. MCAO/R increased cerebral infarct volume and neuronal apoptosis in mice, while 14,15-EET pretreatment increased cerebral infarct volume and neuronal apoptosis. OGD/R induced reactive oxygen species generation, PARP-1 cleavage, and AIF nuclear translocation in cortical neurons. 14,15-EET pretreatment could enhance the antioxidant gene expression of glutathione peroxidase (GSH-Px), heme oxygenase-1 (HO-1) and superoxide dismutase (SOD) in cortical neurons after ischemia and reperfusion. 14,15-EET inhibits the neuronal parthanatos induced by MCAO/R through upregulation of the expression of antioxidant genes and by reducing the generation of reactive oxygen species. This study advances the EET neuroprotection theory and provides a scientific basis for targeted clinical drugs that reduce neuronal parthanatos following cerebral ischemia and reperfusion.


Assuntos
Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Lesões Encefálicas/etiologia , Lesões Encefálicas/metabolismo , Isquemia Encefálica/complicações , Neurônios/efeitos dos fármacos , Parthanatos/efeitos dos fármacos , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/metabolismo , Ácido 8,11,14-Eicosatrienoico/farmacologia , Animais , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/patologia , Isquemia Encefálica/etiologia , Modelos Animais de Doenças , Glucose/metabolismo , Masculino , Camundongos , Modelos Biológicos , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/patologia
12.
Exp Brain Res ; 239(12): 3601-3613, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34591125

RESUMO

The present study aimed to explore the potential mechanism of the effect of hyperbaric oxygenation (HBO) preconditioning on cerebral ischemia and reperfusion injury (CIRI). GSE23160 dataset was used to identify differentially expressed genes (DEGs) from striatum between the middle cerebral artery occlusion (MCAO)/reperfusion and sham rats. The gene clusters with continuous increase and decrease were identified by soft clustering analysis in Mfuzz, and functional enrichment analysis of these genes was performed using clusterProfiler package. The intersection set of the genes with significantly altered expression at post-reperfusion 2, 8, and 24 h were screened in comparison to 0 h (sham group), and the expression of these genes was detected in the MCAO/reperfusion model and HBO preconditioning groups by real-time PCR (RT-PCR) and western blotting. A total of 41 upregulated DEGs, and 7 downregulated DEGs were detected, among which the expression of Gpr84 and Ggta1 was significantly upregulated at each reperfusion phase as compared to the sham group, while the expression of Kcnk3 was significantly downregulated except in the postreperfusion 8 h in the striatum group. RT-PCR and western blotting analyses showed that the expression of Ggta1, Gpr84, and Kcnk3 genes between the MCAO/reperfusion and sham rats were consistent with the bioinformatics analysis. In addition, the HBO preconditioning reduced the expression of Ggta1 and Gpr84 and increased the expression of Kcnk3 in MCAO/reperfusion rats. Kcnk3, Ggta1, and Gpr84 may play a major role in HBO-mediated protection of the brain against CIRI.


Assuntos
Isquemia Encefálica , Oxigenoterapia Hiperbárica , Traumatismo por Reperfusão , Animais , Infarto da Artéria Cerebral Média , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/prevenção & controle
13.
Front Pharmacol ; 12: 791059, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34975487

RESUMO

Background: The dysfunctional blood-brain barrier (BBB)-glymphatic system is responsible for triggering intracerebral amyloid-beta peptide (Aß) accumulation and acts as the key link between ischemic stroke and dementia dominated by Alzheimer's disease (AD). Recently, pyroptosis in cerebral ischemia and reperfusion (I/R) injury is demonstrated as a considerable mechanism causing BBB-glymphatic dysfunctions and Aß acute accumulation in the brain. Targeting glial pyroptosis to protect BBB-glymphatic functions after cerebral I/R could offer a new viewpoint to prevent Aß accumulation and poststroke dementia. Yi-Zhi-Fang-Dai formula (YZFDF) is an herbal prescription used to cure dementia with multiple effects of regulating inflammatory responses and protecting the BBB against toxic Aß-induced damage. Hence, YZFDF potentially possesses neuroprotective effects against cerebral I/R injury and the early pathology of poststroke dementia, which evokes our current study. Objectives: The present study was designed to confirm the potential efficacy of YZFDF against cerebral I/R injury and explore the possible mechanism associated with alleviating Aß acute accumulation. Methods: The models of cerebral I/R injury in rats were built by the method of middle cerebral artery occlusion/reperfusion (MCAO/R). First, neurological function assessment and cerebral infarct measurement were used for confirming the efficacy of YZFDF on cerebral I/R injury, and the optimal dosage (YZFDF-H) was selected to conduct the experiments, which included Western blotting detections of pyroptosis, Aß1-42 oligomers, and NeuN, immunofluorescence observations of glial pyroptosis, aquaporin-4 (AQP-4), and Aß locations, brain water content measurement, SMI 71 (a specific marker for BBB)/AQP-4 immunohistochemistry, and Nissl staining to further evaluate BBB-glymphatic functions and neuronal damage. Results: YZFDF obviously alleviated neurological deficits and cerebral infarct after cerebral I/R in rats. Furthermore, YZFDF could inactivate pyroptosis signaling via inhibiting caspase-1/11 activation and gasdermin D cleavage, ameliorate glial pyroptosis and neuroinflammation, protect against BBB collapse and AQP-4 depolarization, prevent Aß acute accumulation and Aß1-42 oligomers formation, and reduce neuronal damage and increase neurons survival after reperfusion. Conclusion: Our study indicated that YZFDF could exert neuroprotective effects on cerebral I/R injury and prevent Aß acute accumulation in the brain after cerebral I/R associated with inhibiting neuroinflammation-related pyroptosis and BBB-glymphatic dysfunctions.

14.
Neural Regen Res ; 16(6): 1024-1030, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33269746

RESUMO

Apoptosis is an important programmed cell death process involved in ischemia/reperfusion injury. MicroRNAs are considered to play an important role in the molecular mechanism underlying the regulation of cerebral ischemia and reperfusion injury. However, whether miR-670 can regulate cell growth and death in cerebral ischemia/reperfusion and the underlying mechanism are poorly understood. In this study, we established mouse models of transient middle artery occlusion and Neuro 2a cell models of oxygen-glucose deprivation and reoxygenation to investigate the potential molecular mechanism by which miR-670 exhibits its effects during cerebral ischemia/reperfusion injury both in vitro and in vivo. Our results showed that after ischemia/reperfusion injury, miR-670 expression was obviously increased. After miR-670 expression was inhibited with an miR-670 antagomir, cerebral ischemia/reperfusion injury-induced neuronal death was obviously reduced. When miR-670 overexpression was induced by an miR-670 agomir, neuronal apoptosis was increased. In addition, we also found that miR-670 could promote Yap degradation via phosphorylation and worsen neuronal apoptosis and neurological deficits. Inhibition of miR-670 reduced neurological impairments after cerebral ischemia/reperfusion injury. These results suggest that microRNA-670 aggravates cerebral ischemia/reperfusion injury through the Yap pathway, which may be a potential target for treatment of cerebral ischemia/reperfusion injury. The present study was approved by the Institutional Animal Care and Use Committee of China Medical University on February 27, 2017 (IRB No. 2017PS035K).

15.
J Stroke Cerebrovasc Dis ; 29(10): 105126, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32912499

RESUMO

BACKGROUND: Long non-coding RNAs (LncRNAs) have been reported to play important roles in the pathogenesis and development of many diseases, including cerebral ischemia and reperfusion (I/R) injury. In this study, we aimed to investigate the role of LncRNA-Potassium Voltage-Gated Channel Subfamily Q Member 1 opposite strand/antisense transcript 1 (KCNQ1OT1) in cerebral I/R induced neuronal injury, and its underlying mechanisms. METHODS: Primary mouse cerebral cortical neurons treated with oxygen-glucose deprivation and reoxygenation (OGD/R) in vitro and mice subjected to middle cerebral artery occlusion (MCAO) and reperfusion were used to mimic cerebral I/R injury. Small inference RNA (siRNA) was used to knockdown KCNQ1OT1 or microRNA-153-3p (miR-153-3p). Dual-luciferase assay was performed to detect the interaction between KCNQ1OT1 and miR-153-3p and interaction between miR-153-3p and Fork head box O3a (Foxo3). Flow cytometry analysis was performed to detect neuronal apoptosis. qRT-PCR and Western blotting were performed to detect RNA and protein expressions. RESULTS: KCNQ1OT1 and Foxo3 expressions were significantly increased in neurons subjected to I/R injury in vitro and in vivo, and miR-153-3p expression were significantly decreased. Knockdown of KCNQ1OT1 or overexpression of miR-153-3p weakened OGD/R-induced neuronal injury and regulated Foxo3 expressions. Dual-luciferase analysis showed that KCNQ1OT1 directly interacted with miR-153-3p and Foxo3 is a direct target of miR-153-3p. CONCLUSIONS: Our results indicate that LncRNA-KCNQ1OT1 promotes OGD/R-induced neuronal injury at least partially through acting as a competing endogenous RNA (ceRNA) for miR-153-3p to regulate Foxo3a expression, suggesting LncRNA-KCNQ1OT1 as a potential therapeutic target for cerebral I/R injury.


Assuntos
Córtex Cerebral/metabolismo , Proteína Forkhead Box O3/metabolismo , Infarto da Artéria Cerebral Média/terapia , MicroRNAs/metabolismo , Neurônios/metabolismo , RNA Longo não Codificante/metabolismo , Traumatismo por Reperfusão/metabolismo , Reperfusão/efeitos adversos , Animais , Hipóxia Celular , Células Cultivadas , Córtex Cerebral/patologia , Proteína Forkhead Box O3/genética , Regulação da Expressão Gênica , Glucose/deficiência , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , Masculino , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Neurônios/patologia , RNA Longo não Codificante/genética , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/patologia , Transdução de Sinais
16.
Neurochem Res ; 45(8): 1888-1901, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32447509

RESUMO

Selenium has been shown to possess antioxidant and neuroprotective effects by modulating mitochondrial function and activating mitochondrial biogenesis. Our previous study has also suggested that selenium protected neurons against glutamate toxicity and hyperglycemia-induced damage by regulating mitochondrial fission and fusion. However, it is still not known whether the mitochondrial biogenesis is involved in selenium alleviating hyperglycemia-aggravated cerebral ischemia reperfusion (I/R) injury. The object of this study is to define whether selenium protects neurons against hyperglycemia-aggravated cerebral I/R injury by promoting mitochondrial biogenesis. In vitro oxygen deprivation plus high glucose model decreased cell viability, enhanced reactive oxygen species production, and meanwhile stimulated mitochondrial biogenesis signaling. Pretreated with selenium significantly decreased cell death and further activated the mitochondrial biogenesis signaling. In vivo 30 min of middle cerebral artery occlusion in the rats under hyperglycemic condition enhanced neurological deficits, enlarged infarct volume, exacerbated neuronal damage and oxidative stress compared with normoglycemic ischemic rats after 24 h reperfusion. Consistent to the in vitro results, selenium treatment alleviated ischemic damage in hyperglycemic ischemic animals. Furthermore, selenium reduced the structural changes of mitochondria caused by hyperglycemic ischemia and further promoted the mitochondrial biogenesis signaling. Selenium activates mitochondrial biogenesis signaling, protects mitochondrial structure integrity and ameliorates cerebral I/R injury in hyperglycemic rats.


Assuntos
Infarto da Artéria Cerebral Média/prevenção & controle , Mitocôndrias/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Biogênese de Organelas , Selenito de Sódio/uso terapêutico , Animais , Morte Celular/efeitos dos fármacos , Linhagem Celular , Hiperglicemia/fisiopatologia , Infarto da Artéria Cerebral Média/fisiopatologia , Masculino , Camundongos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Transdução de Sinais/efeitos dos fármacos , Superóxido Dismutase/metabolismo , Proteína Desacopladora 2/metabolismo
17.
J Pharmacol Sci ; 143(1): 9-16, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32107104

RESUMO

This study investigated the long-term effects of dihydrocapsaicin (DHC)-induced angiogenesis and improved functional outcomes in cerebral ischemia and reperfusion (I/R) rats. Middle cerebral artery occlusion was induced in I/R rats for 2 h, followed by reperfusion. The animals were divided into three groups: sham, I/R + vehicle, and I/R + DHC (10 mg/kg body weight). Fourteen days after I/R injury, the DHC-treated I/R rats had decreased neurological deficit scores, infarct volume, and brain morphology changes. DHC-induced angiogenesis significantly increased the expression of angiogenic factor proteins, such as hypoxia inducible factor 1α (HIF-1α), vascular endothelial growth factor (VEGF), and matrix metalloprotease 9 (MMP-9), at 3 d and 14 d following I/R and also increased the expression of angiogenic inhibitors, such as angiopoietin 1 (Ang-1) and its receptor tyrosine kinase (Tie-2), at 14 d following reperfusion. DHC-mediated angiogenesis was confirmed by a significant increase in positive BrdU labeling that co-localized with the von Willebrand factor (an endothelial cell marker) at 14 d after I/R. Furthermore, rotarod and pole tests demonstrated that DHC promoted functional recovery when compared with the vehicle group. Thus, the results reveal that DHC mediates angiogenesis and functional recovery after an ischemic stroke.


Assuntos
Indutores da Angiogênese , Isquemia Encefálica/tratamento farmacológico , Capsaicina/análogos & derivados , Neovascularização Fisiológica/efeitos dos fármacos , Recuperação de Função Fisiológica/efeitos dos fármacos , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Encéfalo/irrigação sanguínea , Encéfalo/metabolismo , Encéfalo/patologia , Isquemia Encefálica/patologia , Capsaicina/farmacologia , Capsaicina/uso terapêutico , Modelos Animais de Doenças , Expressão Gênica/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Neovascularização Fisiológica/genética , Ratos Wistar , Traumatismo por Reperfusão/patologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
18.
Biomolecules ; 9(10)2019 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-31547018

RESUMO

Ischemic stroke is a clinically common cerebrovascular disease whose main risks include necrosis, apoptosis and cerebral infarction, all caused by cerebral ischemia and reperfusion (I/R) injury. This process has particular significance for the treatment of stroke patients. Notoginseng leaf triterpenes (PNGL), as a valuable medicine, have been discovered to have neuroprotective effects. However, it was not confirmed that whether PNGL may possess neuroprotective effects against cerebral I/R injury. To explore the neuroprotective effects of PNGL and their underlying mechanisms, a middle cerebral artery occlusion/reperfusion (MCAO/R) model was established. In vivo results suggested that in MCAO/R model rats, PNGL pretreatment (73.0, 146, 292 mg/kg) remarkably decreased infarct volume, reduced brain water content, and improved neurological functions; moreover, PNGL (73.0, 146, 292 mg/kg) significantly alleviated blood-brain barrier (BBB) disruption and inhibited neuronal apoptosis and neuronal loss caused by cerebral I/R injury, while PNGL with a different concertation (146, 292 mg/kg) significantly reduced the concentrations of IL-6, TNF-α, IL-1 ß, and HMGB1 in serums in a dose-dependent way, which indicated that inflammation inhibition could be involved in the neuroprotective effects of PNGL. The immunofluorescence and western blot analysis showed PNGL decreased HMGB1 expression, suppressed the HMGB1-triggered inflammation, and inhibited microglia activation (IBA1) in hippocampus and cortex, thus dose-dependently downregulating inflammatory cytokines including VCAM-1, MMP-9, MMP-2, and ICAM-1 concentrations in ischemic brains. Interestingly, PNGL administration (146 mg/kg) significantly downregulated the levels of p-P44/42, p-JNK1/2 and p-P38 MAPK, and also inhibited expressions of the total NF-κB and phosphorylated NF-κB in ischemic brains, which was the downstream pathway triggered by HMGB1. All of these results indicated that the protective effects of PNGL against cerebral I/R injury could be associated with inhibiting HMGB1-triggered inflammation, suppressing the activation of MAPKs and NF-κB, and thus improved cerebral I/R-induced neuropathological changes. This study may offer insight into discovering new active compounds for the treatment of ischemic stroke.


Assuntos
Infarto Encefálico/prevenção & controle , Proteína HMGB1/metabolismo , Infarto da Artéria Cerebral Média/tratamento farmacológico , Panax notoginseng/química , Traumatismo por Reperfusão/tratamento farmacológico , Triterpenos/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Barreira Hematoencefálica/efeitos dos fármacos , Infarto Encefálico/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/etiologia , Masculino , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Folhas de Planta/química , Ratos , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/etiologia , Transdução de Sinais/efeitos dos fármacos , Triterpenos/farmacologia
19.
Brain Res Bull ; 153: 250-256, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31545998

RESUMO

INTRODUCTION & OBJECTIVE: Cerebral ischemia causes physiological and biochemical cellular changes that ultimately result in structural and functional damage to hippocampal neurons. Ischemia also raises endogenous adenosine release that in turn has neuroprotective effects. The purpose of this study was to evaluate the effect of exogenous adenosine on mitigating neuronal lesions to the CA1 region of hippocampus and A2A protein expression following cerebral I/R in rats. METHODS: Male Wistar rats were randomly assigned to three experimental groups (sham, ischemia + control, and ischemia + adenosine). A daily dose of adenosine (0.1 mg/ml/kg, i.p.) was administered starting 24 h post-ischemia for 7 days. Ischemia was induced by occlusion of both common carotid arteries for 45 min. Cresyl violet and Hematoxylin Eosin staining were used to assess lesion extent and location. To investigate the expression and protein levels, immunohistochemistry and enzyme-linked immunosorbent assay method was used. RESULTS: The cerebral ischemia caused neuronal loss in the CA1 region and reduced sensorimotor functions in lesion animals. Injection of adenosine significantly diminished cell death and improved sensorimotor functional recovery. Moreover, the expression and concentration of A2A protein was significantly greater in the adenosine group compared to the ischemia group. CONCLUSION: This study showed that the administration of exogenous adenosine promotes protection against cell death and supports functional recovery following ischemic injury.


Assuntos
Adenosina/farmacologia , Isquemia Encefálica/tratamento farmacológico , Adenosina/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Isquemia Encefálica/fisiopatologia , Região CA1 Hipocampal/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Modelos Animais de Doenças , Hipocampo/efeitos dos fármacos , Masculino , Neurônios/efeitos dos fármacos , Neuroproteção/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Wistar , Recuperação de Função Fisiológica/efeitos dos fármacos , Traumatismo por Reperfusão/patologia
20.
Biomed Pharmacother ; 118: 109260, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31548176

RESUMO

OBJECTIVE: Cerebral ischemia reperfusion injury (CIRI) is a major cause of ischemic stroke (IS) deterioration. Considering the intricate mechanism of the pathological process of CIRI, most drugs only work on one target. The neurovascular unit (NVU) puts forward the concept of neuroprotection from nerve protection to global stabilization. The NVU plays an important role in maintaining the brain microenvironment. This would promote neuronal survival and overall neurological recovery, which would likely lead to the reduction of mortality rate. Previous studies have shown that 4-methoxy benzyl alcohol (4-MA) ameliorated neurological score and cerebral infarct volume and reduced the concentration of Evans blue (EB) in brain tissue. In this research, we investigated the effects of 4-MA on NVU microenvironment improvement in rats impaired by middle cerebral artery occlusion/reperfusion (MCAO/R). METHODS: First, we established a rat model of middle cerebral artery occlusion (MCAO) so as to use Western blot analysis, immunofluorescence and transmission electron microscopy (TEM) evaluating the NVU's protection of 4-MA. Then we established a primary cortical neuron model of oxygen glucose deprivation and re-oxygenation (OGD/R) with the objective of identifying whether 4-MA exhibited anti-oxidant and anti-apoptotic effects on neurons. RESULTS: NVU ultra structural changes were improved by 4-MA. Immunofluorescence and western blot showed that 4-MA protected NVUs through enhancement of the expression of the symbolic neuronal proteins Microtubule Associated Protein-2(MAP-2), and attenuation of protein expression of Asy symbolic protein Glial Fibrillary Acidic Protein(GFAP). Furthermore, in the OGD/R model of I/R injury in vitro, 4-MA significantly increased Superoxide dismutase(SOD), Nitric Oxide(NO), B-cell lymphoma-2(Bcl-2), decreased Bcl-2-Associated X(Bax) and increased Bcl-2/Bax. CONCLUSION: 4-MA can play the role of anti-ischemic stroke drug by ameliorating the microenvironment of NVUs while its neuroprotective effects will contribute towards the inhibition of the antioxidant and anti-apoptotic activities.


Assuntos
Álcool Benzílico/farmacologia , Fármacos Neuroprotetores/farmacologia , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/metabolismo , Masculino , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/metabolismo , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...