Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Pharmacol ; 12: 722907, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34733156

RESUMO

Mechanical ventilation is an indispensable life-support treatment for acute respiratory failure in critically ill patients, which is generally believed to involve uncontrolled inflammatory responses. Oxytocin (OT) has been reported to be effective in animal models of acute lung injury. However, it is not clear whether Oxytocin has a protective effect on ventilator-induced lung injury (VILI). Therefore, in this study, we aimed to determine whether OT can attenuate VILI and explore the possible mechanism of this protection. To this end, a mouse VILI model was employed. Mice were pretreated with OT 30 min before the intraperitoneal injection of saline or nigericin and ventilation for 4 h, after which they were euthanized. Pathological changes, lung wet/dry (W/D) weight ratio, myeloperoxidase (MPO) activity, the levels of inflammatory cytokines [i.e., interleukin (IL)-1ß, IL-6, and IL-18] in lung tissues and bronchoalveolar lavage fluid (BALF), and expression of NLRP3, Toll-like receptor 4 (TLR4), caspase-1, nuclear factor (NF)-κB, and GSDMD in lung tissues were measured. OT treatment could reduce pathological injury, the W/D ratio, and MPO activity in VILI mice. Our data also indicated that OT administration alleviated the expression of TLR4/My-D88 and the activation of NF-κB, NLRP3, and caspase-1 in lung tissues from the VILI mice model. Furthermore, OT also decreased the levels of IL-1ß, IL-6, and IL-18 in the bronchoalveolar lavage fluid. Moreover, the OT administration may alleviate the activation of GSDMD partially through its effects on the NLRP3-mediated pathway. Collectively, OT exerted a beneficial effect on VILI by downregulating TLR4-and NLRP3-mediated inflammatory pathways.

2.
Neurosci Lett ; 736: 135258, 2020 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-32693014

RESUMO

Ischemia-reperfusion (I/R) injury is accompanied by high mortality and morbidity. Unfortunately, there are few effective therapeutic medicines and strategies to enhance its outcome. Hydroxysafflor Yellow A (HSYA) exerts multiple biological activities and has potential protective effects against I/R injury in the brain, liver and heart. However, its underlying mechanism is still unclear. Here, we investigated whether HSYA modulates apoptosis and neuro-inflammation through the Glycogen synthase kinase-3ß(GSK3ß)-mediated pathway in a transient middle cerebral artery occlusion (MCAO) rat model and oxygen/glucose deprivation (OGD)-challenged primary neuronal cultures both in vivo and in vitro. Male Wistar rats were subjected to MCAO for 2 h, followed by 24 h of reperfusion. HSYA was administered 15 min after occlusion, SB216763 (GSK3ß inhibitor) was injected to the left ventricle of the rat 6 h prior to MCAO. After 24 h of perfusion, apoptosis-associated protein and inflammatory markers were detected by western blotting. Meanwhile, terminal-deoxynucleotidyl transferase mediated nick end labeling(TUNEL) assay was used to evaluate the number of apoptotic cells in OGD-challenged neurons, cleaved caspase-3 were evaluated by Immunofluorescence (IF). Our data indicated that HSYA administration reduced infarct volume, decreased neurological deficit scores, elevated GSK3ß phosphorylation and inhibited the activation of iNOS, NF-κB, and capase-3 in the penumbra of I/R rats. Moreover, blockade of GSK3ß partly reversed the protective effect of HSYA on I/R by regulating NF-κB and caspase-3 both in vivo and in vitro. Collectively, we found that HSYA ameliorates I/R injury through its anti-inflammatory and anti-apoptotic effects via modulation of GSK-3ß phosphorylation.


Assuntos
Isquemia Encefálica/metabolismo , Encéfalo/efeitos dos fármacos , Chalcona/análogos & derivados , Glicogênio Sintase Quinase 3 beta/metabolismo , Quinonas/farmacologia , Traumatismo por Reperfusão/metabolismo , Animais , Apoptose/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Isquemia Encefálica/patologia , Chalcona/farmacologia , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , Masculino , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Wistar , Traumatismo por Reperfusão/patologia , Transdução de Sinais/fisiologia
3.
Pharm Biol ; 58(1): 686-694, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32658590

RESUMO

CONTEXT: Hydroxysafflor yellow A (HSYA) has been shown to have neuroprotective effects in cerebral infarction. However, its underlying roles in apoptosis and inflammation in Parkinson's disease (PD) are unknown. OBJECTIVE: The present study investigates the effects and underlying mechanisms of HSYA on dopaminergic (DA) neurodegeneration, inflammation, and apoptosis. MATERIALS AND METHODS: The PD model was established by 2 µL of 6-hyroxydopamine (6-OHDA) (3 µg/µL) striatal injection in C57BL/6J mice with different doses of HSYA (2, 4, or 8 mg/kg). In vitro, after being treated with HSYA for 1 h, SH-SY5Y cells were exposed to 6-OHDA for 24 h before analysis. Expression of tyrosine hydroxylase (TH) in substantia nigra (SN) and corpus striatum (STR) was evaluated by immunohistochemistry (IHC) and western blot. In addition, apoptosis-related and inflammatory proteins were examined by western blot. RESULTS: Administration of HSYA significantly reduced the Apomorphine (APO)-induced rotation, decreased from 122.5 ± 15.1 (6-OHDA group) to 47.2 ± 14.3 (8 mg/kg HSYA group). HSYA partially restored a deficit in the SN and STR of PD mice brains in TH. Furthermore, western blot analysis revealed that HSYA reduced inflammatory proteins, including iNOS, COX-2 and NF-κB and attenuated the elevation of DA neuronal apoptosis observed in PD. In vitro assays showed that HSYA reduced the levels of p-p38 and p-JNK and increased that of p-ERK in 6-OHDA-leisoned SH-SY5Y cells. CONCLUSIONS: These findings indicate that HSYA protects against 6-OHDA induced DA neurodegeneration partly by regulating the MAPK inflammatory signalling pathway and apoptosis which highlight its therapeutic potential in the treatment of PD.


Assuntos
Chalcona/análogos & derivados , Neurônios Dopaminérgicos/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Transtornos Parkinsonianos/tratamento farmacológico , Quinonas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Chalcona/administração & dosagem , Chalcona/farmacologia , Corpo Estriado/efeitos dos fármacos , Dopamina/metabolismo , Neurônios Dopaminérgicos/patologia , Relação Dose-Resposta a Droga , Inflamação/patologia , Inflamação/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/administração & dosagem , Oxidopamina/toxicidade , Transtornos Parkinsonianos/fisiopatologia , Quinonas/administração & dosagem , Transdução de Sinais/efeitos dos fármacos
4.
Oncol Lett ; 10(3): 1444-1446, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26622687

RESUMO

Giant cystic pheochromocytomas (GPCCs) are rare adrenal tumors and the majority of them present as asymptomatic. As a result GPCCs often remain undiagnosed until surgery and therefore the surgical team face a greater challenge in perioperative management. The present study describes the case of a 36 year-old woman with an undiagnosed GPCC, which was successfully resected despite the occurrence of perioperative cardiovascular events, including hypertension, hypotension, ventricular arrhythmias, acute heart failure, acute myocardial infarction, and the patient was discharged home without any recurrence. It should be considered in retroperitoneal tumour of patients with nonspecific symptoms and given adequate treatment to promote the perioperative safety.

5.
Mol Med Rep ; 12(5): 7039-44, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26299324

RESUMO

Surgical procedures cause a decrease in lymphocyte proliferation rate, an increase in apoptosis and shifts the balance of T­helper (Th)1/Th2 cells towards anti­cell­mediated immunity (CMI) Th2 dominance, which is relevant to the immunosuppressive effects of CMI, postoperative septic complications and the formation of tumor metastasis. Previous studies have revealed that lidocaine exhibits antibacterial actions; regulating inflammatory responses, reducing postoperative pain and affecting the duration spent in hospital. Thus, the present study hypothesized that lidocaine may exert a protective effect on the CMI of patients undergoing surgery for the removal of a primary tumor. A total of 30 adult female patients diagnosed with cervical cancer were recruited to the present study and were randomized into two groups. The lidocaine group received an intravenous bolus dose of 1.5 mg/kg lidocaine, followed by continuous infusion at 1.5 mg/kg/h until discharge from the operating room. The control group received the same volume of normal saline. A 10 ml sample of venous blood was drawn, and the lymphocytes were isolated using Ficoll­paque 1 day prior to surgery, at discharge from the operating room and 48 h post­surgery. The proliferation rate of the lymphocytes was assessed using a Cell Counting Kit­8 assay and was found to be higher in the lidocaine group. The early apoptosis of lymphocytes was attenuated following lidocaine treatment at 48 h post­surgery, as detected using flow cytometry with Annexin V­fluorescein isothiocyanate/propidium iodide staining. The level of interferon (IFN)­Î³ in the serum at 48 h was significantly decreased following surgery in the control group, compared with the pre­surgical values (3.782 ± 0.282, vs. 4.089 ± 0.339 pg/ml, respectively) and the ratio of IFN­Î³ to interleukin­4 was well preserved in the lidocaine group. In conclusion, the present study demonstrated that the intraoperative systemic administration of lidocaine exerted a protective effect on CMI in patients with cervical cancer undergoing radical hysterectomy. This may be beneficial in reducing the occurrence of postoperative septic complications and tumor metastasis formation.


Assuntos
Anestésicos Locais/administração & dosagem , Imunidade Celular/efeitos dos fármacos , Lidocaína/administração & dosagem , Neoplasias do Colo do Útero/cirurgia , Adulto , Idoso , Anestésicos Locais/farmacologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Feminino , Proteína HMGB1/sangue , Humanos , Histerectomia , Injeções Intravenosas , Interferon gama/sangue , Interleucina-4/sangue , Lidocaína/farmacologia , Linfócitos/citologia , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Pessoa de Meia-Idade , Dor Pós-Operatória/prevenção & controle , Estudos Prospectivos , Resultado do Tratamento , Neoplasias do Colo do Útero/tratamento farmacológico , Neoplasias do Colo do Útero/patologia
6.
Int J Clin Exp Med ; 7(9): 3087-95, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25356186

RESUMO

Some studies of animal models of middle cerebral artery occlusion indicate that inflammation plays a key role in the pathogenesis of cerebral ischemia and secondary damage. Flurbiprofen has been suggested to alleviate cerebral ischemia/reperfusion injury in both focal and global cerebral ischemia models, but the mechanisms underlying the protective action are still incompletely understood. In this study we want to investigate the protective effect of flurbiprofen after transient middle cerebral artery occlusion (MCAO) in rats and the role of the NF-κB signaling pathway on this neuroprotective effect. Male Wistar rats were subjected to transient middle cerebral artery occlusion for 2 h, followed by 24 h reperfusion. Flurbiprofen was administrated via tail-vein injection at the onset of reperfusion. HE staining and Immunohistochemistry were carried out to detect the morphological changes in ischemic penumbra cortex. The expression of inflammatory cytokines genes (IL-1ß, IL-6 and TNF-α) and the levels of p-NF-κB (p65) in ischemic penumbra cortex were measured by RT-PCR and western blot. Administration of flurbiprofen at the doses of 5 mg/kg and 10 mg/kg significantly attenuated cerebral ischemia/reperfusion injury, as shown by a reduction in the morphological changes and inhibition of pro-inflammatory cytokine expression in ischemic penumbra cortex. Moreover, our findings further demonstrated that the inhibition of NF-κB activity was involved in the neuroprotective effect of flurbiprofen on inflammatory responses. Flurbiprofen protects against cerebral injury by reducing expression of inflammatory cytokines genes and this effect may be partly due to the inhibition of NF-κB signaling pathway.

7.
Int J Clin Exp Pathol ; 7(7): 3836-44, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25120760

RESUMO

HMGB1 is a necessary and critical mediator of acute lung injury and can act as a chemoattractant and anti-apoptosis factor in injury or repair in diseases. In this study we sought to determine whether HMGB1 is involved in the remodeling of pulmonary artery and investigate the mechanism. A rat model of pulmonary artery remodeling was successful induced with LPS infusion and the increasing of pulmonary arteries media was obviously inhibited in rats treated with thrice inject of HMGB1 neutralizing antibody. The percent of areas of tunica media to total artery wall was (0.53 ± 0.15), (0.81 ± 0.10) and (0.59 ± 0.11) in control, LPS and antibody group respectively (p<0.05). Meanwhile, treatment with HMGB1 neutralizing antibody not only decreased the level of HMGB1 mRNA and protein significantly, but inhibited the expression of PCAN and Bcl-2 as well. On the contrary, Bax, a gen which represented the apoptosis, revealed an absolutely reversed trend to Bcl-2 in pulmonary arteries. Experiments in vitro showed that HMGB1 could stimulate the proliferation of hPASMC in MTT test and increase the number of migrated cells in a concentration-dependent manner in chemotaxis assay using modified Boyden chambers. In conclusion, data from this study support the concept that HMGB1 is involved in the remodeling of pulmonary artery by enhancing proliferation and migration of smooth muscle cell. Inhibiting HMGB1 may be a new target to deal with the remodeling of pulmonary artery.


Assuntos
Proteína HMGB1/metabolismo , Miócitos de Músculo Liso/patologia , Artéria Pulmonar/patologia , Remodelação Vascular/fisiologia , Animais , Western Blotting , Linhagem Celular , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Modelos Animais de Doenças , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Masculino , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real
8.
Neurochem Int ; 66: 27-32, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24462584

RESUMO

CXC195 showed strongest protective effects among the ligustrazine derivatives in cells and prevented apoptosis induced by H2O2 injury. We recently demonstrated that CXC195 protected against cerebral ischemia/reperfusion (I/R) injury by its antioxidant activity. However, whether the anti-apoptotic action of CXC195 is involved in cerebral I/R injury is unknown. Here, we investigated the role of CXC195 in apoptotic processes induced by cerebral I/R and the possible signaling pathways. Male Wistar rats were submitted to transient middle cerebral artery occlusion for 2h, followed by 24h reperfusion. CXC195 was injected intraperitoneally at 2h and 12h after the onset of ischemia. The number of apoptotic cells was measured by TUNEL assay, apoptosis-related protein cleaved caspase-3, Bcl-2, Bax and the phosphorylation levels of Akt and GSK3ß in ischemic penumbra were assayed by western blot. The results showed that administration of CXC195 at the doses of 3mg/kg and 10mg/kg significantly inhibited the apoptosis by decreasing the number of apoptotic cells, decreasing the level of cleaved caspase-3 and Bax, and increasing the level of Bcl-2 in rats subjected to I/R injury. Simultaneously, CXC195 treatment markedly increased the phosphorylation of Akt and GSK3ß. Blockade of PI3K activity by wortmannin, dramatically abolished its anti-apoptotic effect and lowered both Akt and GSK3ß phosphorylation levels. Our study firstly demonstrated that CXC195 protected against cerebral I/R injury by reducing apoptosis in vivo and PI3K/Akt/GSK3ß pathway involved in the anti-apoptotic effect.


Assuntos
Isquemia Encefálica/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Proteína Oncogênica v-akt/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Piperazinas/uso terapêutico , Pirazinas/uso terapêutico , Traumatismo por Reperfusão/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Isquemia Encefálica/prevenção & controle , Glicogênio Sintase Quinase 3 beta , Masculino , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Piperazinas/farmacologia , Pirazinas/farmacologia , Distribuição Aleatória , Ratos , Ratos Wistar , Traumatismo por Reperfusão/prevenção & controle , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
9.
PLoS One ; 8(10): e77909, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24130900

RESUMO

Neuroinflammation is involved in various central nervous system (CNS) disorders, including brain infections, ischemia, trauma, stroke, and degenerative CNS diseases. In the CNS inflammation, secretory phospholipase A2-IIA (sPLA2-IIA) acts as a mediator, resulting in the generation of the precursors of pro-inflammatory lipid mediators, such as prostaglandins (PGs) and leukotrienes (LTs). However, the role of sPLA2-IIA in neuroinflammation is more complicated and remains unclear yet. In the present study, we investigated the effect of sPLA2-IIA inhibition by specific inhibitor SC-215 on the inflammation in LPS-induced mice cerebral cortex and primary astrocytes. Our results showed that the inhibition of sPLA2-IIA alleviated the release of PGE2 by suppressing the activation of ERK1/2, cPLA2α, COX-2 and mPGES-1. These findings demonstrated that sPLA2-IIA showed the potential to regulate the neuroinflammation in vivo and in vitro, indicating that sPLA2-IIA might be a novel target for the treatment of acute neuroinflammation.


Assuntos
Córtex Cerebral/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Fosfolipases A2 do Grupo II/antagonistas & inibidores , Inflamação/prevenção & controle , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fosfolipases A2 Citosólicas/antagonistas & inibidores , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/imunologia , Células Cultivadas , Córtex Cerebral/enzimologia , Córtex Cerebral/imunologia , Fosfolipases A2 do Grupo II/imunologia , Inflamação/enzimologia , Inflamação/imunologia , Lipopolissacarídeos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosfolipases A2 Citosólicas/imunologia , Ratos , Ratos Wistar
10.
Neurochem Res ; 38(11): 2268-75, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23990223

RESUMO

Hydroxysafflor yellow A (HSYA) is the major active chemical component of the flower of the safflower plant, Carthamus tinctorius L. Previously, its neuroprotection against cerebral ischemia-reperfusion (I/R) injury was reported by anti-oxidant action and suppression of thrombin generation. Here, we investigate the role of HSYA in cerebral I/R-mediated apoptosis and possible signaling pathways. Male Wistar rats were subjected to transient middle cerebral artery occlusion for 2 h, followed by 24 h reperfusion. HSYA was administered via tail-vein injection just 15 min after occlusion. The number of apoptotic cells was measured by TUNEL assay, apoptosis-related proteins Bcl-2, Bax and the phosphorylation levels of Akt and GSK3ß in ischemic penumbra were assayed by western blot. The results showed that administration of HSYA at the doses of 4 and 8 mg/kg significantly inhibited the apoptosis by decreasing the number of apoptotic cells and increasing the Bcl-2/Bax ratio in rats subjected to I/R injury. Simultaneously, HSYA treatment markedly increased the phosphorylations of Akt and GSK3ß. Blockade of PI3K activity by wortmannin dramatically abolished its anti-apoptotic effect and lowered both Akt and GSK3ß phosphorylation levels. Taken together, these results suggest that HSYA protects against cerebral I/R injury partly by reducing apoptosis via PI3K/Akt/GSK3ß signaling pathway.


Assuntos
Isquemia Encefálica/prevenção & controle , Chalcona/análogos & derivados , Quinase 3 da Glicogênio Sintase/metabolismo , Infarto da Artéria Cerebral Média/tratamento farmacológico , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinonas/uso terapêutico , Traumatismo por Reperfusão/prevenção & controle , Animais , Apoptose/efeitos dos fármacos , Chalcona/uso terapêutico , Glicogênio Sintase Quinase 3 beta , Masculino , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Proteína X Associada a bcl-2/metabolismo
11.
Biol Pharm Bull ; 36(9): 1420-7, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23803230

RESUMO

The present paper proposes a new approach for fitting the plasma effect-site equilibrium rate constant (Ke0) of propofol to satisfy the condition that the effect-site concentration (Ce) is equal at the time of loss of consciousness (LOC) and recovery of consciousness (ROC). Forty patients receiving intravenous anesthesia were divided into 4 groups and injected propofol 1.4, 1.6, 1.8, or 2 mg/kg at 1,200 mL/h. Durations from the start of injection to LOC and to ROC were recorded. LOC and ROC were defined as an observer's assessment of alertness and sedation scale change from 3 to 2 and from 2 to 3, respectively. Software utilizing bisection method iteration algorithms was built. Then, Ke0 satisfying the CeLOC=CeROC condition was estimated. The accuracy of the Ke0 estimated by our method was compared with the Diprifusor TCI Pump built-in Ke0 (0.26 min(-1)), and the Orchestra Workstation built-in Ke0 (1.21 min(-1)) in another group of 21 patients who were injected propofol 1.4 to 2 mg/kg. Our results show that the population Ke0 of propofol was 0.53 ± 0.18 min(-1). The regression equation for adjustment by dose (mg/kg) and age was Ke0=1.42-0.30 × dose-0.0074 × age. Only Ke0 adjusted by dose and age achieved the level of accuracy required for clinical applications. We conclude that the Ke0 estimated based on clinical signs and the two-point fitting method significantly improved the ability of CeLOC to predict CeROC. However, only the Ke0 adjusted by dose and age and not a fixed Ke0 value can meet clinical requirements of accuracy.


Assuntos
Anestésicos Intravenosos/farmacocinética , Estado de Consciência/efeitos dos fármacos , Propofol/farmacocinética , Adulto , Algoritmos , Período de Recuperação da Anestesia , Anestésicos Intravenosos/administração & dosagem , Anestésicos Intravenosos/sangue , Feminino , Humanos , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade , Propofol/administração & dosagem , Propofol/sangue , Software
13.
Biochem Biophys Res Commun ; 409(4): 808-13, 2011 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-21624354

RESUMO

Apoptosis is one of the major mechanisms of cell death during cerebral ischemia and reperfusion injury. Flurbiprofen has been shown to reduce cerebral ischemia/reperfusion injury in both focal and global cerebral ischemia models, but the mechanism remains unclear. This study aimed to investigate the potential association between the neuroprotective effect of flurbiprofen and the apoptosis inhibiting signaling pathways, in particularly the Akt/GSK-3ß pathway. A focal cerebral ischemia rat model was subjected to middle cerebral artery occlusion (MCAO) for 120 min and then treated with flurbiprofen at the onset of reperfusion. The infarct volume and the neurological deficit scores were evaluated at 24h after reperfusion. Cell apoptosis, apoptosis-related proteins and the levels of p-Akt and p-GSK-3ß in ischemic penumbra were measured using TUNEL and western blot. The results showed that administration of flurbiprofen at the doses of 5 and 10mg/kg significantly attenuated brain ischemia/reperfusion injury, as shown by a reduction in the infarct volume, neurological deficit scores and cell apoptosis. Moreover, flurbiprofen not only inhibited the expression of Bax protein and p-GSK-3ß, but also increased the expression of Bcl-2 protein, the ratio of Bcl-2/Bax as well as the P-Akt level. Taken together, these results suggest that flurbiprofen protects the brain from ischemia/reperfusion injury by reducing apoptosis and this neuroprotective effect may be partly due to the activation of Akt/GSK-3ß signaling pathway.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Isquemia Encefálica/tratamento farmacológico , Flurbiprofeno/uso terapêutico , Quinase 3 da Glicogênio Sintase/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Citoproteção , Glicogênio Sintase Quinase 3 beta , Masculino , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...