Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Neurochem Res ; 49(3): 692-705, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38047987

RESUMO

Narirutin (Nar) is a flavonoid that is abundantly present in citrus fruits and has attracted considerable attention because of its diverse pharmacological activities and low toxicity. Here, we evaluated the preventive effects of Nar in middle cerebral artery occlusion/reperfusion (MCAO/R)-injured mice and oxygen-glucose deprivation/reperfusion (OGD/R)-injured bEnd.3 cells. Pretreatment with Nar (150 mg/kg) for 7 days effectively reduced infarct volume, improved neurological deficits, and significantly inhibited neuronal death in the hippocampus and cortex in MCAO/R-injured mice. Moreover, anti-apoptotic effects of Nar (50 µM) were observed in OGD/R-injured bEnd.3 cells. In addition, Nar pre-administration regulated blood-brain barrier function by increasing tight junction-related protein expression after MCAO/R and OGD/R injury. Nar also inhibited NOD-like receptor protein 3 (NLRP3) inflammasome activation by reducing the expression of thioredoxin-interacting protein (TXNIP) in vivo and in vitro. Taken together, these results provide new evidence for the use of Nar in the prevention and treatment of ischemic stroke.


Assuntos
Isquemia Encefálica , Dissacarídeos , Flavanonas , Traumatismo por Reperfusão , Ratos , Camundongos , Animais , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Proteínas NLR , Ratos Sprague-Dawley , Células Endoteliais/metabolismo , Inflamassomos/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/prevenção & controle , Traumatismo por Reperfusão/metabolismo , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/metabolismo , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/prevenção & controle , Isquemia Encefálica/metabolismo , Proteínas de Ciclo Celular
2.
Mol Med ; 29(1): 44, 2023 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-37013491

RESUMO

BACKGROUND: Dysregulated long non-coding RNAs participate in the development of diabetic cerebral ischemia. This study aimed to investigate the underlying mechanism of lncRNA MALAT1 in diabetic cerebral ischemia. METHOD: Middle cerebral artery occlusion (MCAO) was performed to establish diabetic cerebral I/R in vivo. TTC and neurological deficits assessment were performed to assess cerebral ischemic injury. LDH was conducted to detect cytotoxicity. RT-qPCR and western blotting assays were applied to determine mRNA and protein expression. Flow cytometry was performed to detect the pyroptosis of BV2 cells. Immunofluorescence and FISH were conducted for subcellular localization of MALAT1 and STAT1. ELISA was performed to determine cytokine release. Dual luciferase reporter, RIP, and ChIP assays were used to validate the interaction between STAT1 and MALAT1/NLRP3. Diabetes aggravated cerebral injury in vivo and in vitro. Diabetic cerebral ischemia induced inflammatory response and inflammation-induced cell pyroptosis. RESULT: MALAT1 was overexpressed in diabetic cerebral ischemia models in vivo and in vitro. However, knockdown of MALAT1 suppressed inflammatory response and the pyroptosis of BV2 cells. Moreover, MALAT1 interacted with STAT1 to transcriptionally activate NLRP3. Knockdown of STAT1 significantly reversed the effects of MALAT1. Furthermore, STAT1 promotes the MALAT1 transcription. MALAT1 interacts with STAT1 to promote the pyroptosis of microglias induced by diabetic cerebral ischemia through activating NLRP3 transcription. CONCLUSION: Thus, knockdown of MALAT1 may be a potential promising therapy target for diabetic cerebral ischemia.


Assuntos
Isquemia Encefálica , Diabetes Mellitus , MicroRNAs , RNA Longo não Codificante , Traumatismo por Reperfusão , Isquemia Encefálica/genética , Microglia/metabolismo , MicroRNAs/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Piroptose , Traumatismo por Reperfusão/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT1/farmacologia , Animais
3.
Bioorg Chem ; 132: 106350, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36681044

RESUMO

Oxidative stress has been confirmed to be closely related to the occurrence and development of cerebral ischemic/reperfusion (I/R). The Keap1-Nrf2 pathway is widely recognized as a defensive system to maintain cellular redox homeostasis. Targeting Keap1-Nrf2 interaction by small molecules to release Nrf2 should be a promising strategy to treat cerebral I/R injury. The piperazinyl-naphthalenesulfonamide 6 K was reported to be a Keap1-Nrf2 protein-protein interaction inhibitor, showing promising antioxidative effect. Herein, this study is to investigate whether 6 K could prevent brain from I/R injury. The related mechanism of oxidative stress was also elucidated using in vivo mice middle cerebral artery occlusion (MCAO) model and in vitro SH-SY5Y oxygen-glucose deprivation/reperfusion (OGD/R) model. The results indicated that treatment of 6 K markedly decreased infarct volume, apoptotic neurons and oxidative damage and promoted neurologic recovery in vivo. The cell model revealed that the reactive oxygen species (ROS) was decreased, and cell viability was increased. Western blots and immunofluorescence staining demonstrated that compound treatment promoted Nrf2 release and nuclear translocation. The downstream protective enzymes were significantly enhanced at both in vivo and in vitro levels. Collectively, 6 K is a promising protective agent against cerebral I/R injury through activation of Nrf2 to suppress oxidative stress.


Assuntos
Isquemia Encefálica , Neuroblastoma , Fármacos Neuroprotetores , Traumatismo por Reperfusão , Camundongos , Humanos , Animais , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Fator 2 Relacionado a NF-E2/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch , Estresse Oxidativo , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico
4.
Neurochem Res ; 47(8): 2230-2243, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35482135

RESUMO

Various studies have evidenced the neuroprotective role of PDE4 inhibitors. However, whether PDE4 inhibitor, Piclamilast pharmacological post-treatment is protective during cerebral ischemia reperfusion-induced injury remains unknown. Therefore, this study design included testing the hypothesis that Piclamilast administered at the beginning of a reperfusion phase (Piclamilast pPost-trt) shows protective effects and explores & probes underlying downstream mechanisms. Swiss albino male mice were subjected to global ischemic and reperfusion injury for 17 min. The animals examined cerebral infarct size, biochemical parameters, inflammatory mediators, and motor coordination. For memory, assessment mice were subjected to morris water maze (MWM) and elevated plus maze (EPM) test. Histological changes were assessed using HE staining. Piclamilast pPost-trt significantly reduced I/R injury-induced deleterious effects on biochemical parameters of oxidative stress, inflammatory parameters, infarct size, and histopathological changes, according to the findings. These neuroprotective effects of pPost-trt are significantly abolished by pre-treatment with selective CREB inhibitor, 666-15. Current study concluded that induced neuroprotective benefits of Piclamilast Post-trt, in all probability, maybe mediated through CREB activation. Hence, its neuroprotective effects can be further explored in clinical settings.


Assuntos
Isquemia Encefálica , Fármacos Neuroprotetores , Inibidores da Fosfodiesterase 4 , Traumatismo por Reperfusão , Animais , Benzamidas , Infarto Cerebral/patologia , Camundongos , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Inibidores da Fosfodiesterase 4/farmacologia , Inibidores da Fosfodiesterase 4/uso terapêutico , Piridinas , Traumatismo por Reperfusão/patologia
5.
J Biochem Mol Toxicol ; 36(4): e22982, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34978116

RESUMO

Ischemic stroke is one of the main causes of death and disability. Circular RNAs (circRNAs) have received extensive attention in the pathogenesis of ischemic stroke. Here, we evaluated the role of circCDC14A in cerebral ischemia-reperfusion (CI/R) injury in vivo and in vitro. The expression of circCDC14A was significantly upregulated in the middle cerebral artery occlusion (MCAO) model and oxygen and glucose deprivation/reoxygenation (OGD/R)-treated HT22 cells. Knockdown of circCDC14A suppressed the cell viability reduction caused by OGD/R, as well as cell damage and apoptosis. Mechanistically, circCDC14A acted as a sponge for miR-23a-3p and promoted the expression of chemokine stromal-derived factor-1 (CXCL12) by negatively regulating miR-23a-3p. Rescue experiments further confirmed that miR-23a-3p inhibitor or circCDC14A-overexpression vectors blocked the beneficial effects of circCDC14A knockdown in OGD/R-induced HT22 cells. Moreover, knockdown of circCDC14A suppressed MCAO-induced cerebral infarction and neurological damage, as well as the brain tissue damage and neuronal apoptosis in vivo. Consistently, miR-23a-3p antagomir treatment abolished the cerebral protective effects of circCDC14A knockdown on MCAO mice. In conclusion, circCDC14A promoted CI/R injury by regulating the miR-23a-3p/CXCL12 axis, which suggested that circCDC14A may become a potential therapeutic target for CI/R injury.


Assuntos
Isquemia Encefálica , AVC Isquêmico , MicroRNAs , Traumatismo por Reperfusão , Animais , Apoptose , Isquemia Encefálica/genética , Glucose , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Circular/genética , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/prevenção & controle
6.
J Proteome Res ; 19(3): 1154-1168, 2020 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-31940440

RESUMO

Diosgenin (DIO), the starting material for the synthesis of steroidal anti-inflammatory drugs in the pharmaceutical industry, has been previously demonstrated to display pharmaceutical effects against cerebral ischemic reperfusion (I/R). However, the alterations of brain proteome profiles underlying this treatment remain elusive. In the present study, the proteomics analysis of the brain tissues from I/R rats after DIO treatment was performed using an integrated TMT-based quantitative proteomic approach coupled with the liquid chromatography with tandem mass spectrometry technology. A total of 5043 proteins (ProteomeXchange identifier: PXD016303) were identified, of which 58 common differentially expressed proteins were significantly dysregulated in comparison between sham versus I/R and I/R versus DIO. The eight validated proteins including EPG5, STAT2, CPT1A, EIF2AK2, GGCT, HIKESHI, TNFAIP8, and EMC6 by quantitative polymerase chain reaction and western blotting consistently supported the TMT-based proteomic results, which were mainly associated with autophagy and inflammation response. Considering the anti-inflammatory characters of DIO, the biological functions of STAT2 and HIKESHI that are the probable direct anti-inflammatory targets were further investigated during the course of I/R treated with DIO. In addition, the combination of verified STAT2 and HIKESHI in peripheral blood samples from stroke patients resulted in the area under the curve value of 0.765 with P < 0.004 to distinguish stroke patients from healthy controls. Taken together, the current findings first mapped comprehensive proteomic changes after I/R was treated with DIO to better decipher the molecular mechanisms mainly based on the anti-inflammatory aspect underlying this therapeutic effect, providing a foundation for developing potentially therapeutic targets of anti-I/R of DIO and clinically prognostic biomarkers of stroke.


Assuntos
Isquemia Encefálica , Diosgenina , Traumatismo por Reperfusão , Animais , Encéfalo , Isquemia Encefálica/tratamento farmacológico , Proteínas de Transporte , Diosgenina/farmacologia , Humanos , Proteínas de Membrana , Proteômica , Ratos , Reperfusão , Traumatismo por Reperfusão/tratamento farmacológico
7.
Biochem Biophys Res Commun ; 531(4): 622-627, 2020 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-32819716

RESUMO

Liposomal fasudil as a treatment for cerebral ischemia/reperfusion (I/R) injury has been demonstrated to be effective in animal models due to the high accumulation of liposomes in damaged brain tissue. However, it is still unclear what effect drug release rate has on the treatment of I/R injury, where pathology progresses dramatically in a short time. In the present study, we assessed four formulations of liposomal fasudil. The results of an in vitro drug release assay showed that the release properties of fasudil were changed by varying the lipid composition and internal phase of the liposomes. Based on these results, differences in the transition of fasudil plasma concentration were monitored after the administration of each type of liposomal fasudil in normal rats. A pharmacokinetic study showed that higher levels of drug retention in liposomal fasudil resulted in higher fasudil plasma concentration. Finally, treatment of I/R injury model rats with liposomal fasudil revealed that a mid-level release rate of fasudil from liposomes resulted in the greatest therapeutic effect among the formulations. In conclusion, these results demonstrate that an optimized drug release rate from liposomes enhances the therapeutic effect of fasudil for the treatment of cerebral I/R injury.


Assuntos
1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , Lipossomos/química , Traumatismo por Reperfusão/tratamento farmacológico , 1,2-Dipalmitoilfosfatidilcolina/química , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/sangue , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacocinética , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Sulfato de Amônio/química , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Ácido Cítrico/química , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Composição de Medicamentos , Sistemas de Liberação de Medicamentos/métodos , Liberação Controlada de Fármacos , Lipossomos/farmacocinética , Masculino , Fosfatidilcolinas/química , Compostos de Amônio Quaternário/química , Ratos Wistar , Traumatismo por Reperfusão/patologia , Resultado do Tratamento
8.
J Dairy Sci ; 103(2): 1151-1163, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31837800

RESUMO

This study aimed to investigate the modulation activity of heated and nonheated lactoferrins in an inflammatory pathway in anoxia and reoxygenation cell and cerebral ischemic reperfusion mouse models. Rat pheochromocytoma 12 (PC-12) cells were subjected to oxygen and glucose deprivation in vitro to construct an anoxia and reoxygenation cell model, and Institute for Cancer Research (ICR) mice were given carotid artery "ligation-relaxation" in vivo to construct a cerebral ischemic reperfusion mouse model. The protein levels of toll-like receptor 4 (TLR-4) and downstream inflammatory proteins including nuclear factor-κB (NF-κB), tumor necrosis factor-α (TNF-α), and IL-1ß were detected. Meanwhile, metabonomic detection of overall metabolites of PC-12 cells was performed to screen out the specific changed metabolite affected by lactoferrin at the condition of anoxia and reoxygenation. The results showed that lactoferrin could inhibit the TLR-4-related pathway triggered by anoxia and reoxygenation and ischemic reperfusion. A total of 41 significantly changed metabolites were identified by metabonomic analysis, and glutathione was seen as a metabolite of interest in suppressing TLR-4-related pathway in anoxia and reoxygenation cell models. However, heated lactoferrin lost the ability of attenuating the TLR-4-related pathway. The loss of modulation activity of heated lactoferrin might be due to its protein aggregation, which was evidenced by larger average particle diameter than the unheated lactoferrin. This study is the first to investigate the effect of heat treatment on the modulation activity of lactoferrin in the TLR-4-related pathway in anoxia and reoxygenation cell and cerebral ischemic reperfusion mouse models, and indicate that lactoferrin may serve as a dietary intervention for cerebral ischemia.


Assuntos
Isquemia Encefálica/metabolismo , Hipóxia Celular , Hipóxia-Isquemia Encefálica/metabolismo , Lactoferrina/farmacologia , Receptor 4 Toll-Like/metabolismo , Animais , Isquemia Encefálica/prevenção & controle , Modelos Animais de Doenças , Glucose/farmacologia , Lactoferrina/química , Masculino , Camundongos , Camundongos Endogâmicos ICR , NF-kappa B/metabolismo , Oxigênio/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos , Temperatura , Fator de Necrose Tumoral alfa/metabolismo
9.
Biol Pharm Bull ; 41(4): 575-584, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29607930

RESUMO

Previous reports have indicated that isosteviol sodium (STVNa) has neuroprotective effects against acute focal cerebral ischemia in rats; however, the exact underlying mechanisms and ideal treatment paradigm are not known. To find a reasonable method for STVNa administration and to determine its possible therapeutic mechanisms, we characterized the protective effects of single-dose and multiple-dose STVNa in cerebral ischemic/reperfusion (I/R) injury in rats. Single and multiple treatments with 10 mg/kg STVNa were administered intraperitoneally after injury to investigate its neuroprotective effects. Neurobehavioral deficits and infarct volume were assessed 7 d after ischemia. Both STVNa treatments reduced infarct volumes, improved neurological behaviors, preserved cellular morphology, enhanced neuronal survival, and suppressed cell apoptosis. Multiple treatments performed better than single treatment. Reactive astrogliosis was apparent at 7 d after injury and was significantly inhibited by multiple STVNa treatments but not single treatment. These results indicate that STVNa exerts neuroprotection by different mechanisms in the acute and delayed phases of I/R. Specifically, STVNa neuroprotection in the delayed phase of injury was found to be accompanied with the inhibition of astrogliosis.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Diterpenos do Tipo Caurano/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Isquemia Encefálica/patologia , Masculino , Neurônios/efeitos dos fármacos , Neurônios/patologia , Ratos Sprague-Dawley , Traumatismo por Reperfusão/patologia
10.
Zhongguo Zhong Yao Za Zhi ; 42(11): 2139-2145, 2017 Jun.
Artigo em Chinês | MEDLINE | ID: mdl-28822160

RESUMO

To observe the effect of active components group of Xiaoxuming decoction (XXMD) on brain mitochondria in cerebral ischemia/reperfusion rats during early recovery period, and study its protective mechanism for nerves in cerebral ischemia/reperfusion rats during early recovery period. Cerebral ischemia model of middle cerebral artery occlusion in rats was established by suture method, and reperfusion was conducted 2 h later. The degree of cerebral ischemia in rats was evaluated by using Zea-Longa's standard grading method, and the model rats were randomly divided into model group, Xiaoxuming decoction active components low, medium and high dose groups and positive drug Ginaton group, with sham operated rats as control group. Gradient centrifugation was used to extract the mitochondria from rat brain after 5 days of drug administration. Then the mitochondrial respiratory function was measured by Clark oxygen electrode method; mitochondrial membrane potential and the mitochondrial reactive oxygen species(ROS) level were detected by fluorescence probe methods; and the activity of mitochondrial succinodehydrogenase (SDH) and the content of ATP in the ischemic region of MCAO rats were measured by spectrophotometric method. The results showed that as compared with the model group, XXMD could significantly improve mitochondrial respiratory activity, increase the activity of SDH, reduce the level of ROS, increase mitochondrial membrane potential and obviously promote the synthesis of ATP in brain tissues. The results indicated that XXMD active components group could alleviate the energy metabolism disorders, protect brain mitochondrial damage and improve mitochondrial function in MCAO rats, which may be the mechanism of its neuroprotection activity.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Medicamentos de Ervas Chinesas/farmacologia , Mitocôndrias/efeitos dos fármacos , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Encéfalo/efeitos dos fármacos , Infarto da Artéria Cerebral Média , Potencial da Membrana Mitocondrial , Ratos , Espécies Reativas de Oxigênio/metabolismo
11.
Microvasc Res ; 99: 36-42, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25708051

RESUMO

Cerebral ischemic-reperfusion injury as an inflammatory and microcirculation dysfunction pathological condition negatively affects the clinical outcome of stroke patients. The novel inflammatory procoagulant protein fgl2 has been reported to play a role in some inflammatory and coagulation dysregulation diseases. This study aimed to examine the relationship between fgl2 expression and infarct size in an acute cerebral ischemic-reperfusion rat model. We studied fgl2 mRNA and protein expressions in cerebral tissue and peripheral macrophages, and the expressions of several inflammatory factors (TNF-α, IL-1ß, MCP-1, and IL-8) in serum samples from rats with acute cerebral ischemic-reperfusion injury. Fiber microthrombosis in situ contributed to the microvascular thrombosis in acute cerebral ischemic-reperfusion injury, and fgl2 expression tended to strongly correlate with cerebral infarct size. The expression levels of the other inflammatory factors significantly increased but weakly correlated with cerebral infarct size. These findings support the potential of fgl2 level as a novel biomarker of acute cerebral ischemic-reperfusion injury.


Assuntos
Biomarcadores/metabolismo , Regulação da Expressão Gênica , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Traumatismo por Reperfusão/patologia , Animais , Quimiocina CCL2/metabolismo , Citocinas/sangue , Fibrinogênio/metabolismo , Inflamação , Interleucina-1beta/metabolismo , Interleucina-8/metabolismo , Macrófagos/metabolismo , Masculino , Microcirculação , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Trombose/fisiopatologia , Fator de Necrose Tumoral alfa/metabolismo
13.
J Ethnopharmacol ; 332: 118321, 2024 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-38735418

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Cordyceps has a long medicinal history as a nourishing herb in traditional Chinese medicine (TCM). Ischemic cardio-cerebrovascular diseases (CCVDs), including cerebral ischemic/reperfusion injury (CI/RI) and myocardial ischemic/reperfusion injury (MI/RI), are major contributors to mortality and disability in humans. Numerous studies have indicated that Cordyceps or its artificial substitutes have significant bioactivity on ischemic CCVDs, however, there is a lack of relevant reviews. AIM OF THE STUDY: This review was conducted to investigate the chemical elements, pharmacological effects, clinical application and drug safety of Cordycepson ischemic CCVDs. MATERIALS AND METHODS: A comprehensive search was conducted on the Web of Science, PubMed, Chinese National Knowledge Infrastructure (CNKI), and Wanfang databases using the keywords "Cordyceps", "Cerebral ischemic/reperfusion injury", and "Myocardial ischemic/reperfusion injury" or their synonyms. The retrieved literature was then categorized and summarized. RESULTS: The study findings indicated that Cordyceps and its bioactive components, including adenosine, cordycepin, mannitol, polysaccharide, and protein, have the potential to protect against CI/RI and MI/RI by improving blood perfusion, mitigating damage from reactive oxygen species, suppressing inflammation, preventing cellular apoptosis, and promoting tissue regeneration. Individually, Cordyceps could reduce neuronal excitatory toxicity and blood-brain barrier damage caused by cerebral ischemia. It can also significantly improve cardiac energy metabolism disorders and inhibit calcium overload caused by myocardial ischemia. Additionally, Cordyceps exerts a significant preventive or curative influence on the factors responsible for heart/brain ischemia, including hypertension, thrombosis, atherosclerosis, and arrhythmia. CONCLUSION: This study demonstrates Cordyceps' prospective efficacy and safety in the prevention or treatment of CI/RI and MI/RI, providing novel insights for managing ischemic CCVDs.


Assuntos
Cordyceps , Humanos , Cordyceps/química , Animais , Medicina Tradicional Chinesa/métodos , Isquemia Encefálica/tratamento farmacológico
14.
Zhen Ci Yan Jiu ; 49(4): 391-397, 2024 Apr 25.
Artigo em Inglês, Chinês | MEDLINE | ID: mdl-38649207

RESUMO

OBJECTIVES: To observe the effect of electroacupuncture (EA) at "Baihui" (GV20) and "Shenting" (GV24) on the rats' behavior and the transforming precursor of brain-derived neurotrophic factor (proBDNF) into mature brain-derived neurotrophic factor (mBDNF) in the hippocampus of rats with learning and memory impairment induced by cerebral ischemia-reperfusion (IR), so as to explore its mechanisms underlying improvement of learning and memory ability. METHODS: SD rats were randomly divided into blank, sham operation, model, and EA groups, with 6 rats in each group. The model of IR was established by occlusion of the middle cerebral artery. EA (1 Hz/20 Hz) was applied to GV24 and GV20 for 30 min, once daily for 14 days. The neurological function was evaluated according to the Zea Longa's score criteria 24 h after modeling and after intervention. Morris water maze test was used to detect the learning and memory function of the rats. TTC staining was used to evaluate the cerebral infarction volume on the affected side. The protein expression levels of proBDNF, mBDNF, tissue plasminogen activator (tPA), tyrosine kinase receptor B (TrkB) and p75 neurotrophin receptor (p75NTR) in hippocampal tissue were detected by Western blot. RESULTS: Compared with the sham operation group, the neurological function score, the percentage of cerebral infarction volume and the expression levels of proBDNF and p75NTR protein in hippocampus were increased (P<0.01), while the times of crossing the original platform and the total distance in the target quadrant, the expression levels of mBDNF, TrkB and tPA protein and the ratio of mBDNF/proBDNF were decreased (P<0.01, P<0.05) in the model group. Compared with the model group, the neurological function score, the percentage of cerebral infarction volume, and the expression levels of proBDNF and p75NTR protein in hippocampus were decreased (P<0.01, P<0.05), while the times of crossing the original platform, the total distance in the target quadrant, and the expression levels of mBDNF, TrkB and tPA protein and the ratio of mBDNF/proBDNF were increased (P<0.05, P<0.01) in the EA group. CONCLUSIONS: EA can alleviate learning and memory impairment in IR rats, which may be related to its function in up-regulating the expression of tPA protein and promoting the transformation of proBDNF to mBDNF, thus improving the synaptic plasticity.


Assuntos
Isquemia Encefálica , Fator Neurotrófico Derivado do Encéfalo , Eletroacupuntura , Transtornos da Memória , Plasticidade Neuronal , Precursores de Proteínas , Traumatismo por Reperfusão , Animais , Humanos , Masculino , Ratos , Pontos de Acupuntura , Isquemia Encefálica/metabolismo , Isquemia Encefálica/terapia , Isquemia Encefálica/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Fator Neurotrófico Derivado do Encéfalo/genética , Hipocampo/metabolismo , Aprendizagem , Memória , Transtornos da Memória/terapia , Transtornos da Memória/metabolismo , Transtornos da Memória/etiologia , Ratos Sprague-Dawley , Receptor trkB/metabolismo , Receptor trkB/genética , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/terapia , Traumatismo por Reperfusão/genética
15.
Neurochem Int ; 169: 105586, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37442439

RESUMO

Cerebral ischemic/reperfusion (I/R) injury has become a great challenge harming patients' life. This study aims to explore the regulatory role of Preso during cerebral I/R injury and to elucidate the potential mechanism. Here, we established a middle cerebral artery occlusion/reperfusion (MCAO/IR) rat model and an oxygen-glucose deprivation/reoxygenation (OGD/R)-mediated PC12 cell model to evaluate the expression and role of Preso following cerebral I/R injury. Histopathological injury and infarct size were assessed by hematoxylin and eosin (HE) and 2,3,5-Triphenyltertrazolium chloride (TTC) staining. Double immunofluorescence staining was performed to assess neuronal apoptosis in brain tissues. Cell counting kit-8 (CCK-8) and flow cytometry were performed to evaluate cell viability and apoptosis, respectively. The reactive oxygen species (ROS) and nitric oxide (NO) levels were detected using their respective detection kits, and the expression of corresponding proteins was examined adopting Western blot. The results showed that Preso was upregulated in OGD/R-induced PC12 cells and MCAO rats. Preso knockdown significantly reduced OGD/R-caused viability loss, apoptosis and oxidative stress in PC12 cells, and reduced infarct size, attenuated histological injury, and inhibited apoptosis and oxidative stress in the brain tissues from MCAO rats, as well as inhibiting the expression of postsynaptic density protein-95 (PSD95) and nitric oxide synthase (nNOS) and repressing YAP phosphorylation in vitro. In addition, the protective role of Preso knockdown against cerebral I/R injury was partly strengthened by IC87201, the nNOS/PSD95 interaction inhibitor, or weakened by Verteporfin (Vert), an inhibitor of YAP. In conclusion, Perso knockdown might exert a protective role against cerebral I/R injury via regulating PSD95-nNOS and YAP pathways, providing a potential therapeutic target for the treatment of ischemic stroke.


Assuntos
Isquemia Encefálica , Traumatismo por Reperfusão , Acidente Vascular Cerebral , Animais , Ratos , Apoptose , Isquemia Encefálica/metabolismo , Proteína 4 Homóloga a Disks-Large/metabolismo , Regulação para Baixo , Infarto da Artéria Cerebral Média/metabolismo , Óxido Nítrico Sintase/metabolismo , Reperfusão , Traumatismo por Reperfusão/metabolismo , Acidente Vascular Cerebral/metabolismo , Proteínas de Sinalização YAP/metabolismo
16.
PeerJ ; 11: e14483, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36643627

RESUMO

Background: Icaritin (ICT) has been previously demonstrated to display protective effects against cerebral ischemic reperfusion (I/R) by inhibiting oxidative stress, but the mechanism remains unclear. This study aimed to explore the mechanism from the perspective of metabolomics. Methods: A mice cerebral artery occlusion/reperfusion (MCAO/R) model was explored to mimic cerebral ischemic reperfusion and protective effect of ICT was assessed by neurologic deficit scoring, infarct volume and brain water content. Ultra-high-performance liquid chromatography electrospray ionization orbitrap tandem mass spectrometry (UHPLC-ESI-QE-Orbitrap-MS) based metabolomic was performed to explore potential biomarkers. Brain tissue metabolic profiles were analyzed and metabolic biomarkers were identified through multivariate data analysis. The protein levels of Nrf2, HO-1 and HQO1 were assayed by western blot. The release of malondialdehyde (MDA) and the activity of superoxide dismutase (SOD), glutathione peroxidase (GSH-Px) and catalase (CAT) were detected using corresponding assay kits. Results: The results showed that after ICT treatment, the neurological deficit, cerebral infarction area, brain edema and the level of MDA in brain tissue of MCAO/R mice were significantly reduced. Meanwhile, ICT enhanced the activity of SOD, CAT and GSH-Px. Western blot results confirmed that ICT up-regulated the protein levels of antioxidant-related protein including Nrf2, HO-1 and NQO1. According to the metabolomic profiling of brain tissues, clear separations were observed among the Sham, Model and ICT groups. A total of 44 biomarkers were identified, and the identified biomarkers were mainly related to linoleic acid metabolism, arachidonic acid metabolism, alanine, aspartate and glutamate metabolism, arginine biosynthesis, arginine and proline metabolism, D-glutamine and D-glutamate metabolism, taurine and hypotaurine metabolism and purine metabolism, respectively. At the same time, the inhibitory effect of ICT on arachidonic acid and linoleic acid in brain tissue, as well as the promoting effect on taurine, GABA, NAAG, may be the key factors for the anti-neurooxidative function of mice after MCAO/R injury. Conclusion: Our results demonstrate that ICT has benefits for MCAO/R injury, which are partially related to the suppression of oxidative stress via stimulating the Nrf2 signaling and regulating the production of arachidonic acid, linoleic acid, taurine, GABA, NAAG in brain tissue.


Assuntos
Antioxidantes , Traumatismo por Reperfusão , Animais , Camundongos , Antioxidantes/farmacologia , Ácido Araquidônico , Cromatografia Líquida de Alta Pressão , Ácido gama-Aminobutírico , Ácido Linoleico , Fator 2 Relacionado a NF-E2 , Reperfusão , Traumatismo por Reperfusão/tratamento farmacológico , Superóxido Dismutase/metabolismo , Taurina
17.
Brain Res Bull ; 194: 23-34, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36681251

RESUMO

Mitochondrial dysfunction and endoplasmic reticulum (ER) stress occur in ischemic stroke. The disruption of these two organelles can directly lead to cell death through various signaling pathways. Thus, investigation of the associated molecular mechanisms in cerebral ischemia is a prerequisite for stroke treatment. Pleckstrin homology-like domain family A member 1 (PHLDA1) is a multifunctional protein that can modulate mitochondrial function and ER stress in cardiomyocyte and cancer cells. This work studied the role of PHLDA1 in cerebral ischemic/reperfusion (I/R) injury and explored the underlying mechanisms associated with mitochondrial functions and ER stress. Middle cerebral artery occlusion/reperfusion (MCAO/R)-treated mice and oxygen-glucose deprivation/reoxygenation (OGD/R)-stimulated neurons were used as I/R models in vivo and in vitro, respectively. PHLDA1 was upregulated in ischemic penumbra of MCAO/R-induced mice and OGD/R-exposed neurons. In vitro, PHLDA1 knockdown protected neurons from OGD/R-induced apoptosis. In vivo, PHLDA1 silencing facilitated functional recovery and reduced cerebral infarct volume. Mechanistically, PHLDA1 knockdown promoted PPARγ nuclear translocation, which may mediate the effects on reversion of mitochondrial functions and alleviation of ER stress. In summary, PHLDA1 knockdown alleviates neuronal ischemic injuries in mice. PPARγ activation and mitochondrial dysfunction and endoplasmic reticulum stress attenuation are involved in the underlying mechanisms.


Assuntos
Isquemia Encefálica , Traumatismo por Reperfusão , Animais , Camundongos , Apoptose , Isquemia Encefálica/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Infarto da Artéria Cerebral Média/metabolismo , Mitocôndrias/metabolismo , Neurônios/metabolismo , PPAR gama/metabolismo , Traumatismo por Reperfusão/metabolismo
18.
Antioxidants (Basel) ; 12(5)2023 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-37237865

RESUMO

Cerebral ischemia/reperfusion causes exacerbated neuronal damage involving excessive neuroinflammation and oxidative stress. ROS is considered a signal molecule to activate NLRP3; thus, the ROS/NLRP3/pyroptosis axis plays a vital role in the pathogenesis of cerebral ischemia/reperfusion injury (CIRI). Therefore, targeting the inhibition of the ROS/NLRP3/pyroptosis axis may be a promising therapeutic tactic for CIRI. Epimedium (EP) contains many active ingredients (ICA, ICS II, and ICT), which have a wide range of pharmacological activities. However, whether EP can protect against CIRI remains unknown. Thus, in this study, we designed to investigate the effect and possible underlying mechanism of EP on CIRI. The results showed that treatment with EP dramatically mitigated brain damage in rats following CIRI, which was achieved by suppressing mitochondrial oxidative stress and neuroinflammation. Furthermore, we identified the ROS/NLRP3/pyroptosis axis as a vital process and NLRP3 as a vital target in EP-mediated protection. Most interestingly, the main compounds of EP directly bonded with NLRP3, as reflected by molecular docking, which indicated that NLRP3 might be a promising therapeutic target for EP-elicited cerebral protection. In conclusion, our findings illustrate that ICS II protects against neuron loss and neuroinflammation after CIRI by inhibiting ROS/NLRP3-mediated pyroptosis.

19.
Mol Neurobiol ; 60(10): 5655-5671, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37335462

RESUMO

Paired immunoglobulin-like receptor B (PirB) was identified as a myelin-associated inhibitory protein (MAIP) receptor that plays a critical role in axonal regeneration, synaptic plasticity and neuronal survival after stroke. In our previous study, a transactivator of transcription-PirB extracellular peptide (TAT-PEP) was generated that can block the interactions between MAIs and PirB. We found that TAT-PEP treatment improved axonal regeneration, CST projection and long-term neurobehavioural recovery after stroke through its effects on PirB-mediated downstream signalling. However, the effect of TAT-PEP on the recovery of cognitive function and the survival of neurons also needs to be investigated. In this study, we investigated whether pirb RNAi could alleviate neuronal injury by inhibiting the expression of PirB following exposure to oxygen-glucose deprivation (OGD) in vitro. In addition, TAT-PEP treatment attenuated the volume of the brain infarct and promoted the recovery of neurobehavioural function and cognitive function. This study also found that TAT-PEP exerts neuroprotection by reducing neuronal degeneration and apoptosis after ischemia-reperfusion injury. In addition, TAT-PEP improved neuron survival and reduced lactate dehydrogenase (LDH) release in vitro. Results also showed that TAT-PEP reduced malondialdehyde (MDA) levels, increased superoxide dismutase (SOD) activity and reduced reactive oxygen species (ROS) accumulation in OGD-injured neurons. The possible mechanism was that TAT-PEP could contribute to the damage of neuronal mitochondria and affect the expression of cleaved caspase 3, Bax and Bcl-2. Our results suggest that PirB overexpression in neurons after ischaemic-reperfusion injury induces neuronal mitochondrial damage, oxidative stress and apoptosis. This study also suggests that TAT-PEP may be a potent neuroprotectant with therapeutic potential for stroke by reducing neuronal oxidative stress, mitochondrial damage, degeneration and apoptosis in ischemic stroke.


Assuntos
Isquemia Encefálica , Disfunção Cognitiva , Traumatismo por Reperfusão , Acidente Vascular Cerebral , Humanos , Transativadores/metabolismo , Neurônios/metabolismo , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/metabolismo , Peptídeos/farmacologia , Oxigênio/metabolismo , Proteínas da Mielina/metabolismo , Apoptose , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/metabolismo , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/metabolismo , Isquemia Encefálica/complicações , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/metabolismo
20.
Life Sci ; 313: 121276, 2023 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-36496032

RESUMO

AIM: LncRNAs are highly expressed in the CNS and regulate pathophysiological processes. However, the potential role of lncRNAs inischemic stroke (IS) remains unknown. In this study, we investigated the functions and possible molecular mechanism of lncRNA paternal expressed gene 11 antisense (PEG11as) in this process. METHODS: Middle cerebral artery occlusion/reperfusion (MCAO/R) mice model and N2a cells model from oxygen-glucose deprivation/reoxygenation (OGD/R) were used to simulate cerebral I/R in vivo and in vitro. High-throughput sequencing (RNA-Seq) was used todetect differential expression of lncRNAs in cerebral I/R. QRT-PCR was used to detect the expression of PEG11as and miR-342-5p. Bioinformatics analysis, FISH, luciferase reporter assay, RIP, Western blot, and immunofluorescence were used to detect the interaction between PEG11as, miR-342-5p and PFN1. The effect on neuronal apoptosis was analyzed using loss-of-function combined with TUNEL, Hoechst, and caspase3 activity assays. KEY FINDINGS: 254 lncRNAs were differentially expressed in MCAO1h/R6h mice. Among them, PEG11as was significantly up-regulated. PEG11as down-regulated could markedly attenuate the brain infarct volume, alleviate neurological deficit in vivo, and effectively promote neuron survival, attenuate neuronal apoptosis both in vivo and in vitro. FISH assay discovered that PEG11as was mainly located in the cytoplasm. Furthermore, we demonstrated that PEG11as was able to bind miR-342-5p to inhibit miR-342-5p activity, whereas the down-regulated of miR-342-5p resulted in profilin 1 (PFN1) overexpression and thus promoting apoptosis. SIGNIFICANCE: This study suggests that PEG11as regulates neuronal apoptosis by miR-342-5p/PFN1 axis, which may contribute to our understanding of pathogenesis and provide a potential therapeutic option for cerebral I/R.


Assuntos
Isquemia Encefálica , AVC Isquêmico , MicroRNAs , RNA Longo não Codificante , Traumatismo por Reperfusão , Acidente Vascular Cerebral , Camundongos , Animais , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Profilinas , MicroRNAs/genética , MicroRNAs/metabolismo , Acidente Vascular Cerebral/genética , Traumatismo por Reperfusão/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Apoptose/genética , Glucose/metabolismo , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa