Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Biochem Biophys Res Commun ; 609: 84-92, 2022 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-35421633

RESUMO

Autophagy is a double-edged sword that affects tumor progression by promoting cell survival or death depending on different living contexts. The concrete mechanism by which autophagy modulates the efficacy of radiotherapy for prostate cancer (PC) remains unclear. We exposed RM-1 PC cells to X-ray and explored the role of autophagy in radiation injury. Our results showed increased apoptosis and autophagy levels in RM-1 cells after radiation. Pharmacological inhibition of autophagy by chloroquine significantly mitigated radiation-induced apoptosis, while the enhancement of autophagy by rapamycin aggravated apoptosis. Sirt1, a member of sirtuin family, deacetylates various transcription factors to trigger cell survival in response to radiation injury. We found that radiation led to Sirt1 downregulation, which was reversed by the inhibition of autophagy. On the contrary, enhanced autophagy further diminished protein level of Sirt1. Notably, overexpression of Sirt1 by plasmid significantly alleviated radiation-induced apoptosis, but silenced Sirt1 by siRNA further induced apoptosis, indicating the radioprotective effect of Sirt1 on RM-1 cells. In summary, our findings suggested that autophagy-mediated Sirt1 downregulation might be a promising therapeutic target for PC.


Assuntos
Neoplasias da Próstata , Lesões por Radiação , Sirtuína 1/metabolismo , Animais , Apoptose , Autofagia , Regulação para Baixo , Humanos , Masculino , Camundongos , Neoplasias da Próstata/genética , Neoplasias da Próstata/radioterapia , Tolerância a Radiação , Sirtuína 1/genética
2.
ACS Chem Neurosci ; 15(11): 2223-2232, 2024 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-38634698

RESUMO

Neuronal death resulting from ischemic stroke is the primary cause of adult mortality and disability, and effective neuroprotective agents for poststroke intervention are still lacking. Remote ischemic postconditioning (RIPostC) has demonstrated significant protective effects against ischemia in various organs; however, the specific mechanisms are not fully understood. This study investigated the potential neuroprotective mechanisms of RIPostC in the context of ischemic stroke. Using a rat model of middle cerebral artery occlusion, we found that RIPostC mitigated neurological damage, improved movement in the open-field test, and protected against neuronal apoptosis. In terms of energy metabolism, RIPostC enhanced ATP levels, suppressed lactate content, and increased the production of ketone bodies (KBs). In the ferroptosis assay, RIPostC protected against lipoperoxidation, reversed the reduction of glutathione peroxidase 4 (GPX4), and mitigated the excessive expression of long-chain acyl-CoA synthetase family member 4 (ACSL4). In oxygen-glucose deprivation/reoxygenation-treated HT22 cells, KBs maintained GPX4 levels, suppressed ACSL4 expression, and preserved the mitochondrial cristae number. However, the effect of KBs on the expression of GPX4, ACSL4, and the number of mitochondrial cristae was blocked by erastin. Moreover, both RIPostC and KBs reduced total iron and ferrous ion content by repressing iron transporters both in vitro and in vivo. In conclusion, KBs-induced mitigation of ferroptosis could represent a new therapeutic mechanism for RIPostC in treating stroke.


Assuntos
Coenzima A Ligases , Ferroptose , Infarto da Artéria Cerebral Média , Pós-Condicionamento Isquêmico , Corpos Cetônicos , Neuroproteção , Ferroptose/fisiologia , Animais , Ratos , Pós-Condicionamento Isquêmico/métodos , Corpos Cetônicos/metabolismo , Masculino , Coenzima A Ligases/metabolismo , Neuroproteção/fisiologia , Ratos Sprague-Dawley , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Camundongos , Fármacos Neuroprotetores/farmacologia , AVC Isquêmico/metabolismo , Acidente Vascular Cerebral/metabolismo , Neurônios/metabolismo
3.
Mol Neurobiol ; 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38767837

RESUMO

Cerebral ischemia-reperfusion injury produces excessive reactive oxygen and nitrogen species, including superoxide, nitric oxide, and peroxynitrite (ONOO-). We recently developed a new ONOO--triggered metal-free carbon monoxide donor (PCOD585), exhibiting a notable neuroprotective outcome on the rat middle cerebral artery occlusion model and rendering an exciting intervention opportunity toward ischemia-induced brain injuries. However, its therapeutic mechanism still needs to be addressed. In the pharmacological study, we found PCOD585 inhibited neuronal Bcl2/Bax/caspase-3 apoptosis pathway in the peri-infarcted area of stroke by scavenging ONOO-. ONOO- scavenging further led to decreased Acyl-CoA synthetase long-chain family member 4 and increased glutathione peroxidase 4, to minimize lipoperoxidation. Additionally, the carbon monoxide release upon the ONOO- reaction with PCOD585 further inhibited the neuronal Iron-dependent ferroptosis associated with ischemia-reperfusion. Such a synergistic neuroprotective mechanism of PCOD585 yields as potent a neuroprotective effect as Edaravone. Additionally, PCOD585 penetrates the blood-brain barrier and reduces the degradation of zonula occludens-1 by inhibiting matrix metalloproteinase-9, thereby protecting the integrity of the blood-brain barrier. Our study provides a new perspective for developing multi-functional compounds to treat ischemic stroke.

4.
Neural Regen Res ; 18(5): 1040-1045, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36254990

RESUMO

Ischemic accumulation of succinate causes cerebral damage by excess production of reactive oxygen species. However, it is unknown whether ischemic accumulation of succinate affects neural stem cell proliferation. In this study, we established a rat model of cerebral ischemia/reperfusion injury by occlusion of the middle cerebral artery. We found that succinate levels increased in serum and brain tissue (cortex and hippocampus) after ischemia/reperfusion injury. Oxygen-glucose deprivation and reoxygenation stimulated primary neural stem cells to produce abundant succinate. Succinate can be converted into diethyl succinate in cells. Exogenous diethyl succinate inhibited the proliferation of mouse-derived C17.2 neural stem cells and increased the infarct volume in the rat model of cerebral ischemia/reperfusion injury. Exogenous diethyl succinate also increased the succinylation of the Rho family GTPase Cdc42 but repressed Cdc42 GTPase activity in C17.2 cells. Increasing Cdc42 succinylation by knockdown of the desuccinylase Sirt5 also inhibited Cdc42 GTPase activity in C17.2 cells. Our findings suggest that ischemic accumulation of succinate decreases Cdc42 GTPase activity by induction of Cdc42 succinylation, which inhibits the proliferation of neural stem cells and aggravates cerebral ischemia/reperfusion injury.

5.
Imeta ; 2(3): e116, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38867934

RESUMO

Depression is a common and debilitating condition for which effective treatments are needed. Lepidium meyenii Walp (Maca) is a plant with potential medicinal effects in treating depression. Recently, there has been growing interest in plant-derived extracellular vesicles (EVs) due to their low toxicity and ability to transport to human cells. Targeting the gut-brain axis, a novel strategy for depression management, may be achieved through the use of Maca-derived EVs (Maca-EVs). In this study, we successfully isolated Maca-EVs using gradient ultracentrifugation and characterized their shape, size, and markers (CD63 and TSG101). The in vivo imaging showed that the Dil-labeled Maca-EVs crossed the brain-blood barrier and accumulated in the brain. The behavioral tests revealed that Maca-EVs dramatically recovered the depression-like behaviors of unpredictable chronic mild stress (UCMS) mice. UCMS mice fecal were characterized by an elevated abundance of g_Enterococcus, g_Lactobacillus, and g_Escherichia_Shigella, which were significantly restored by administration of Maca-EVs. The effects of Maca-EVs on the altered microbial and fecal metabolites in UCMS mice were mapped to biotin, pyrimidine, and amino acid (tyrosine, alanine, aspartate, and glutamate) metabolisms, which were closely associated with the serotonin (5-HT) production. Maca-EVs were able to increase serum monoamine neurotransmitter levels in UCMS mice, with 5-HT showing the most significant changes. We further demonstrated that 5-HT improved the expression of brain-derived neurotrophic factor, a key regulator of neuronal plasticity, and its subsequent activation of TrkB/p-AKT signaling by regulating the GTP-Cdc42/ERK pathway. These findings suggest that Maca-EVs enhance 5-HT release, possibly by modulating the gut-brain axis, to improve depression behavior. Our study sheds light on a novel approach to depression treatment using plant-derived EVs.

6.
Front Cardiovasc Med ; 9: 864188, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35509278

RESUMO

Thoracic radiotherapy patients have higher risks of developing radiation-induced heart disease (RIHD). Ionizing radiation generates excessive reactive oxygens species (ROS) causing oxidative stress, while Momordica. charantia and its extract have antioxidant activity. Plant-derived extracellular vesicles (EVs) is emerging as novel therapeutic agent. Therefore, we explored the protective effects of Momordica. charantia-derived EVs-like nanovesicles (MCELNs) against RIHD. Using density gradient centrifugation, we successfully isolated MCELNs with similar shape, size, and markers as EVs. Confocal imaging revealed that rat cardiomyocytes H9C2 cells internalized PKH67 labeled MCELNs time-dependently. In vitro assay identified that MCELNs promoted cell proliferation, suppressed cell apoptosis, and alleviated the DNA damage in irradiated (16 Gy, X-ray) H9C2 cells. Moreover, elevated mitochondria ROS in irradiated H9C2 cells were scavenged by MCELNs, protecting mitochondria function with re-balanced mitochondria membrane potential. Furthermore, the phosphorylation of ROS-related proteins was recovered with increased ratios of p-AKT/AKT and p-ERK/ERK in MCELNs treated irradiated H9C2 cells. Last, intraperitoneal administration of MCELNs mitigated myocardial injury and fibrosis in a thoracic radiation mice model. Our data demonstrated the potential protective effects of MCELNs against RIHD. The MCELNs shed light on preventive regime development for radiation-related toxicity.

7.
Front Pharmacol ; 13: 908830, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35814200

RESUMO

Plant exosome-like nanoparticles (ELNs) have shown great potential in treating tumor and inflammatory diseases, but the neuroprotective effect of plant ELNs remains unknown. In the present study, we isolated and characterized novel ELNs from Momordica charantia (MC) and investigated their neuroprotective effects against cerebral ischemia-reperfusion injury. In the present study, MC-ELNs were isolated by ultracentrifugation and characterized. Male Sprague-Dawley rats were subjected to middle cerebral artery occlusion (MCAO) and MC-ELN injection intravenously. The integrity of the blood-brain barrier (BBB) was examined by Evans blue staining and with the expression of matrix metalloproteinase 9 (MMP-9), claudin-5, and ZO-1. Neuronal apoptosis was evaluated by TUNEL and the expression of apoptotic proteins including Bcl2, Bax, and cleaved caspase 3. The major discoveries include: 1) Dil-labeled MC-ELNs were identified in the infarct area; 2) MC-ELN treatment significantly ameliorated BBB disruption, decreased infarct sizes, and reduced neurological deficit scores; 3) MC-ELN treatment obviously downregulated the expression of MMP-9 and upregulated the expression of ZO-1 and claudin-5. Small RNA-sequencing revealed that MC-ELN-derived miRNA5266 reduced MMP-9 expression. Furthermore, MC-ELN treatment significantly upregulated the AKT/GSK3ß signaling pathway and attenuated neuronal apoptosis in HT22 cells. Taken together, these findings indicate that MC-ELNs attenuate ischemia-reperfusion-induced damage to the BBB and inhibit neuronal apoptosis probably via the upregulation of the AKT/GSK3ß signaling pathway.

8.
Front Cell Neurosci ; 16: 841544, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35308117

RESUMO

Blood-brain barrier (BBB) dysfunction causing edema and hemorrhagic transformation is one of the pathophysiological characteristics of stroke. Protection of BBB integrity has shown great potential in improving stroke outcome. Here, we assessed the efficacy of exosomes extracted from healthy rat serum in protection against ischemic stroke in vivo and in vitro. Exosomes were isolated by gradient centrifugation and ultracentrifugation and exosomes were characterized by transmission electron microscopy (TEM) and nanoparticle tracking video microscope. Exosomes were applied to middle cerebral artery occlusion (MCAO) rats or brain microvascular endothelial cell line (bEnd.3) subjected to oxygen-glucose deprivation (OGD) injury. Serum-derived exosomes were injected intravenously into adult male rats 2 h after transient MCAO. Infarct volume and gross cognitive function were assessed 24 h after reperfusion. Poststroke rats treated with serum-derived exosomes exhibited significantly reduced infarct volumes and enhanced neurological function. Apoptosis was assessed via terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick-end labeling (TUNEL) staining and the expression of B-cell lymphoma-2 (Bcl-2), Bax, and cleaved caspase-3 24 h after injury. Our data showed that serum exosomes treatment strikingly decreased TUNEL+ cells in the striatum, enhanced the ratio of Bcl-2 to Bax, and inhibited cleaved caspase-3 production in MCAO rats and OGD/reoxygenation insulted bEnd.3 cells. Under the consistent treatment, the expression of microtubule-associated protein 1 light chain 3B-II (LC3B-II), LC3B-I, and Sequestosome-1 (SQSTM1)/p62 was detected by Western blotting. Autolysosomes were observed via TEM. We found that serum exosomes reversed the ratio of LC3B-II to LC3B-I, prevented SQSTM1/p62 degradation, autolysosome formation, and autophagic flux. Together, these results indicated that exosomes isolated from healthy serum provided neuroprotection against experimental stroke partially via inhibition of endothelial cell apoptosis and autophagy-mediated BBB breakdown. Intravenous serum-derived exosome treatment may, therefore, provide a novel clinical therapeutic strategy for ischemic stroke.

9.
J Recept Signal Transduct Res ; 31(6): 402-7, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22060185

RESUMO

In this study, we investigated the neuroprotective effects of paclitaxel in transient cerebral ischemia and possible regulatory mechanism of these neuroprotection. Our data showed that paclitaxel can down-regulate the increased MLK3, JNK3, c-Jun, Bcl-2, and caspase-3 phosphorylation induced by ischemia injury. Cresyl violet staining and immunohistochemistry results demonstrated that paclitaxel had neuroprotective effect against ischemia/reperfusion-induced neuronal cell death. These results indicated that paclitaxel has neuroprotection in ischemic injury through JNK3 signaling pathway and provided a novel possible drug in therapeutics of brain ischemia.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Isquemia Encefálica/enzimologia , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Fármacos Neuroprotetores/farmacologia , Paclitaxel/farmacologia , Traumatismo por Reperfusão/enzimologia , Moduladores de Tubulina/farmacologia , Animais , Caspase 3/metabolismo , Morte Celular , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , MAP Quinase Quinase Quinase 11 Ativada por Mitógeno
10.
J Recept Signal Transduct Res ; 29(1): 38-43, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19519168

RESUMO

Here we examined the effects of ischemia preconditioning and ketamine, an NMDA receptor antagonist, on the activation and its nucleus translocation of ERK5 in hippocampal CA1 region. Our results showed ERK5 was not activated in rat hippocampus CA1 region. But in cytosol extracts preconditioned with 3 min of sublethal ischaemia, ERK5 activation was enhanced significantly, with two peaks occurring at 3 hr and 3 days, respectively. This activation returned to base level 3 days later. The results lead us to conclude that preconditioning increased the activations of ERK5 during reperfusion after lethal ischemia through NMDA receptor. Preconditioning increased the activation and nucleus translocation of ERK5 during reperfusion after lethal ischemia through the NMDA receptor. These findings might provide some clues to understanding the mechanism underlying ischemia tolerance and to finding clinical therapies for stroke using the endogenous neuroprotection.


Assuntos
Antagonistas de Aminoácidos Excitatórios/metabolismo , Isquemia/metabolismo , Precondicionamento Isquêmico , Ketamina/metabolismo , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Transdução de Sinais/fisiologia , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Ativação Enzimática , Hipocampo/citologia , Hipocampo/metabolismo , Masculino , Proteína Quinase 7 Ativada por Mitógeno/genética , Ratos , Ratos Sprague-Dawley
11.
Curr Neuropharmacol ; 15(1): 134-156, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27334020

RESUMO

Tissue plasminogen activator (t-PA) is the only FDA-approved drug for acute ischemic stroke treatment, but its clinical use is limited due to the narrow therapeutic time window and severe adverse effects, including hemorrhagic transformation (HT) and neurotoxicity. One of the potential resolutions is to use adjunct therapies to reduce the side effects and extend t-PA's therapeutic time window. However, therapies modulating single target seem not to be satisfied, and a multitarget strategy is warranted to resolve such complex disease. Recently, large amount of efforts have been made to explore the active compounds from herbal supplements to treat ischemic stroke. Some natural compounds revealed both neuro- and bloodbrain- barrier (BBB)-protective effects by concurrently targeting multiple cellular signaling pathways in cerebral ischemia-reperfusion injury. Thus, those compounds are potential to be one-drug-multi-target agents as combined therapy with t-PA for ischemic stroke. In this review article, we summarize current progress about molecular targets involving in t-PA-mediated HT and neurotoxicity in ischemic brain injury. Based on these targets, we select 23 promising compounds from currently available literature with the bioactivities simultaneously targeting several important molecular targets. We propose that those compounds merit further investigation as combined therapy with t-PA. Finally, we discuss the potential drawbacks of the natural compounds' studies and raise several important issues to be addressed in the future for the development of natural compound as an adjunct therapy.


Assuntos
Produtos Biológicos/uso terapêutico , Acidente Vascular Cerebral/terapia , Terapia Trombolítica/métodos , Ativador de Plasminogênio Tecidual/uso terapêutico , Animais , Ácido Ascórbico , Isquemia Encefálica/complicações , Quimioterapia Combinada , Flavonoides , Humanos , Acidente Vascular Cerebral/etiologia , Resultado do Tratamento
12.
Brain Res Bull ; 124: 123-8, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27091695

RESUMO

S-nitrosylation, the nitric oxide-derived post-translational modification of proteins, plays critical roles in various physiological and pathological functions. In this present study, a rat model of cerebral ischemia and reperfusion by four-vessel occlusion was generated to assess MKK4 S-nitrosylation. Immunoprecipitation and immunoblotting were performed to evaluate MKK4 S-nitrosylation and phosphorylation. Neuronal loss was observed using histological detection. These results indicated that endogenous NO promoted the S-nitrosylation of MKK4. However, application of the exogenous NO donor S-nitrosoglutathione (GNSO), an inhibitor of the neuronal nitric oxide synthase 7-nitroindazole (7-NI), and the N-methyl-d-aspartate receptor (NMDAR) antagonist MK801 diminished I/R-induced S-nitrosylation and phosphorylation. These compounds also markedly decreased cerebral I/R-induced degeneration and death of neurons in hippocampal CA1 region in rats. Taken together, we demonstrated for the first time, that cerebral ischemia/reperfusion can induce S-nitrosylation of MKK4. We also found that inhibiting S-nitrosylation and activation of MKK4 resulted in marked decreases in neuronal degeneration and apoptosis, potentially via NMDAR-mediated mechanisms. These findings may lead to a new field of inquiry to investigate the underlying pathogenesis of stoke and the development of novel treatment strategies.


Assuntos
Isquemia Encefálica/patologia , Região CA1 Hipocampal/patologia , MAP Quinase Quinase 4/metabolismo , Neurônios/efeitos dos fármacos , Reperfusão , S-Nitrosoglutationa/uso terapêutico , Animais , Isquemia Encefálica/tratamento farmacológico , Caspase 3/metabolismo , Modelos Animais de Doenças , Maleato de Dizocilpina/uso terapêutico , Indazóis/uso terapêutico , Masculino , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
13.
Sci Rep ; 6: 29246, 2016 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-27385592

RESUMO

It has been demonstrated that Src could modulate NMDA receptor, and PAR1 could also affect NMDAR signaling. However, whether PAR1 could regulate NMDAR through Src under ICH has not yet been investigated. In this study, we demonstrated the role of Src-PSD95-GluN2A signaling cascades in rat ICH model and in vitro thrombin challenged model. Using the PAR1 agonist SFLLR, antagonist RLLFS and Src inhibitor PP2, electrophysiological analysis showed that PAR1 regulated NMDA-induced whole-cell currents (INMDA) though Src in primary cultured neurons. Both in vivo and in vitro results showed the elevated phosphorylation of tyrosine in Src and GluN2A and enhanced interaction of the Src-PSD95-GluN2A under model conditions. Treatment with the PAR1 antagonist RLLFS, AS-PSD95 (Antisense oligonucleotide against PSD95) and Src inhibitor PP2 inhibited the interaction among Src-PSD95-GluN2A, and p-Src, p-GluN2A. Co-application of SFLLR and AS-PSD95, PP2, or MK801 (NMDAR inhibitor) abolished the effect of SF. In conclusion, our results demonstrated that activated thrombin receptor PAR1 induced Src activation, enhanced the interaction among Src-PSD95-GluN2A signaling modules, and up-regulated GluN2A phosphorylation after ICH injury. Elucidation of such signaling cascades would possibly provide novel targets for ICH treatment.


Assuntos
Hemorragia Cerebral/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Quinases da Família src/metabolismo , Animais , Masculino , Neurônios/metabolismo , Fosforilação/fisiologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia , Trombina/metabolismo , Ativação Transcricional/fisiologia , Tirosina/metabolismo , Regulação para Cima/fisiologia
14.
CNS Neurosci Ther ; 21(7): 585-90, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25996167

RESUMO

AIM: Hemorrhagic transformation (HT) is a major complication of delayed tissue plasminogen activator (t-PA) treatment in ischemic stroke. We aimed to explore whether peroxynitrite decomposition catalyst (PDC) could prevent such complication. METHODS: Male Sprague-Dawley (SD) rats were subjected to middle cerebral artery occlusion (MCAO) with t-PA (10 mg/kg) or t-PA plus FeTMPyP (3 mg/kg, a representative PDC) at MCAO for 2 or 5 h and reperfusion for 22 or 19 h, respectively. HT was assessed with hemoglobin assay. Neurological deficit was evaluated with Modified Neurological Severity Score (mNSS). Peroxynitrite formation was examined by detecting 3-nitrotyrosine (3-NT) formation. The expression and activity of MMP-9/MMP-2 were assessed by Western blotting and gelatin zymography. RESULTS: t-PA treatment at 2 h of MCAO did not induce HT but attenuated neurological deficit, whereas treatment at 5 h significantly induced HT and worsened the neurological outcome. Such complications were prevented by FeTMPyP cotreatment. Early t-PA treatment inhibited 3-NT and MMP-9/MMP-2 expression, whereas delayed treatment induced 3-NT and MMP-9/MMP-2 expression and activity. FeTMPyP cotreatment downregulated 3-NT and inhibited MMP-9/MMP-2 in both time points. CONCLUSION: Peroxynitrite decomposition catalyst could prevent hemorrhagic transformation and improve neurological outcome ischemic rat brains with delayed t-PA treatment via inhibiting peroxynitrite-mediated MMP activation.


Assuntos
Fibrinolíticos/efeitos adversos , Hemorragia/induzido quimicamente , Hemorragia/tratamento farmacológico , Hemostáticos/uso terapêutico , Ácido Peroxinitroso/uso terapêutico , Ativador de Plasminogênio Tecidual/efeitos adversos , Animais , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Infarto da Artéria Cerebral Média/tratamento farmacológico , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Doenças do Sistema Nervoso/etiologia , Ratos , Ratos Sprague-Dawley , Reperfusão , Índice de Gravidade de Doença
15.
Brain Res ; 1398: 86-93, 2011 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-21172324

RESUMO

The present study investigates the role of protein kinase C alpha/delta (PKCα/PKCδ) in brain injury induced by intracerebral hemorrhage (ICH) by utilizing a rat model that received intracerebral injections of autologous blood and thrombin (TM). The activation and expression of PKC and PKCδ were analyzed by Western blot and immunohistochemistry. A PKC inhibitor, dihydrochloride (H7), was administrated intraperitoneally after injury to evaluate the effect of inhibition of PKC on ICH and TM induced brain damage. Our data indicate that both ICH and TM increased the expression of PKCα/PKCδ in the brain tissue, and PKCα expression peaked at 6h, while PKCδ expression reached its maximum value at 72h post-injury. Administration of H7 significantly reduced the inflammatory cells infiltrate, permeability of brain-blood barrier (BBB), brain edema, and neuronal death. We conclude that both PKCα and PKCδ play important roles in ICH and TM-induced brain injury, and dihydrochloride (H7) can attenuate brain damage after ICH.


Assuntos
Dano Encefálico Crônico/enzimologia , Dano Encefálico Crônico/fisiopatologia , Hemorragia Cerebral/enzimologia , Hemorragia Cerebral/fisiopatologia , Proteína Quinase C-alfa/fisiologia , Proteína Quinase C-delta/fisiologia , Transdução de Sinais/fisiologia , Trombina/toxicidade , Animais , Dano Encefálico Crônico/patologia , Hemorragia Cerebral/patologia , Modelos Animais de Doenças , Masculino , Proteína Quinase C-alfa/antagonistas & inibidores , Proteína Quinase C-delta/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Trombina/metabolismo
16.
Brain Res ; 1328: 162-70, 2010 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-20197063

RESUMO

Over-activation of NMDA receptor has been widely believed to be the main signal resulting in ischemic cell injury. We recently reported that the triplicate complex NR2A-PSD-95-Src is a signaling module to facilitate NMDA receptor over-activation. In addition, over-activation of NMDA receptor can activate another signaling molecule nNOS, which is also mediated by PSD-95 after cerebral ischemia. Here, we examined whether overexpression of the PDZ1 domain of PSD-95 could disrupt the functional interaction between NMDA receptor and PSD-95 in rat hippocampal CA1 region, and whether or not it could exert a neuroprotective effect against cerebral ischemia. Our results showed that overexpression of PDZ1 domain not only decreased the assembly of NR2A-PSD-95-Src signaling module and the auto-phosphorylation of Src, which mediates NMDA receptor phosphorylation, but also enhanced nNOS (Ser847) phosphorylation. Most importantly, overexpression of PDZ1 domain protected rat hippocampal CA1 neurons against cerebral ischemia injury. These results suggest that overexpression of the PDZ1 domain can perturb the binding of PSD-95 to NMDA receptor, suppress the activity of both NMDA receptor and nNOS, and thus have a neuroprotective effect.


Assuntos
Isquemia Encefálica/metabolismo , Citoproteção/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Quinases da Família src/metabolismo , Sequência de Aminoácidos/fisiologia , Animais , Isquemia Encefálica/fisiopatologia , Domínio Catalítico/fisiologia , Proteína 4 Homóloga a Disks-Large , Regulação para Baixo/fisiologia , Hipocampo/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/química , Proteínas de Membrana/química , Neurônios/metabolismo , Óxido Nítrico/biossíntese , Fosforilação/fisiologia , Estrutura Terciária de Proteína/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/metabolismo , Serina/metabolismo , Transdução de Sinais/fisiologia , Regulação para Cima/fisiologia
17.
Brain Res ; 1276: 151-8, 2009 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-19406109

RESUMO

In this study we investigated whether ethanol could play neuroprotective effects against ischemic brain injury and the related mechanism. Cresyl violet staining results demonstrated that moderate dose of ethanol administered intracerebroventricularly (i.c.v.) had neuroprotective effect against ischemia-reperfusion induced neuronal cell death. Ethanol also inhibited the phosphorylation of JNK3 induced by cerebral ischemia-reperfusion. Three separate drugs, NS3763 (the selective antagonist of GluR5), GluR5 antisense oligodeoxynucleotides (AS-ODNs) and Bicuculline (an antagonist of GABA receptors), were found to inhibit the neuroprotective effect of ethanol. Moreover, the GABA receptor agonist muscimol could attenuate the JNK3 phosphorylation. Taken together, the results suggest that during ischemia-reperfusion ethanol may activate presynaptic GluR5-KA and postsynaptic GABA receptors continuously, and the activation of GABA receptors inhibits the JNK3 signal pathway. The results show a novel potential mechanism underlying ethanol protective effects.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Etanol/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Receptores de GABA/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Benzamidas/administração & dosagem , Bicuculina/administração & dosagem , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Isquemia Encefálica/complicações , Morte Celular/efeitos dos fármacos , GABAérgicos/administração & dosagem , Masculino , Proteína Quinase 10 Ativada por Mitógeno/metabolismo , Muscimol/administração & dosagem , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Oligodesoxirribonucleotídeos Antissenso/administração & dosagem , Ratos , Ratos Sprague-Dawley , Receptores de Ácido Caínico/antagonistas & inibidores , Receptores de Ácido Caínico/genética
18.
Neural Regen Res ; 20(3): 845-857, 2025 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38886957

RESUMO

JOURNAL/nrgr/04.03/01300535-202503000-00029/figure1/v/2024-06-17T092413Z/r/image-tiff It has been shown clinically that continuous removal of ischemia/reperfusion-induced reactive oxygen species is not conducive to the recovery of late stroke. Indeed, previous studies have shown that excessive increases in hypochlorous acid after stroke can cause severe damage to brain tissue. Our previous studies have found that a small amount of hypochlorous acid still exists in the later stage of stroke, but its specific role and mechanism are currently unclear. To simulate stroke in vivo, a middle cerebral artery occlusion rat model was established, with an oxygen-glucose deprivation/reoxygenation model established in vitro to mimic stroke. We found that in the early stage (within 24 hours) of ischemic stroke, neutrophils produced a large amount of hypochlorous acid, while in the recovery phase (10 days after stroke), microglia were activated and produced a small amount of hypochlorous acid. Further, in acute stroke in rats, hypochlorous acid production was prevented using a hypochlorous acid scavenger, taurine, or myeloperoxidase inhibitor, 4-aminobenzoic acid hydrazide. Our results showed that high levels of hypochlorous acid (200 µM) induced neuronal apoptosis after oxygen/glucose deprivation/reoxygenation. However, in the recovery phase of the middle cerebral artery occlusion model, a moderate level of hypochlorous acid promoted the proliferation and differentiation of neural stem cells into neurons and astrocytes. This suggests that hypochlorous acid plays different roles at different phases of cerebral ischemia/reperfusion injury. Lower levels of hypochlorous acid (5 and 100 µM) promoted nuclear translocation of ß-catenin. By transfection of single-site mutation plasmids, we found that hypochlorous acid induced chlorination of the ß-catenin tyrosine 30 residue, which promoted nuclear translocation. Altogether, our study indicates that maintaining low levels of hypochlorous acid plays a key role in the recovery of neurological function.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA