Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell Physiol Biochem ; 35(1): 315-25, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25591773

RESUMO

BACKGROUND: Previous studies have shown ketamine can alter the proliferation and differentiation of neural stem cells (NSCs) in vitro. However, these effects have not been entirely clarified in vivo in the subventricular zone (SVZ) of neonatal rats. The present study was designed to investigate the effects of ketamine on the proliferation and differentiation of NSCs in the SVZ of neonatal rats in vivo. METHODS: Postnatal day 7 (PND-7) male Sprague-Dawley rats were administered four injections of 40 mg/kg ketamine at 1-h intervals, and then 5-bromodeoxyuridine (BrdU) was injected intraperitoneally at PND-7, 9 and 13. NSC proliferation was assessed with Nestin/BrdU double-labeling immunostaining. Neuronal and astrocytic differentiation was evaluated with ß-tubulin III/BrdU and GFAP/BrdU double-labeling immunostaining, respectively. The expressions of nestin, ß-tubulin III and GFAP were measured using Western blot analysis. The apoptosis of NSCs and astrocytes in the SVZ of neonatal rats was evaluated using nestin/caspase-3 and GFAP/caspase-3 double-labeling immunostaining. RESULTS: Neonatal ketamine exposure significantly reduced the number of nestin/BrdU and GFAP/BrdU double-positive cells in the SVZ. Meanwhile, the expressions of nestin and GFAP in the SVZ from the ketamine group were significantly decreased compared those in the control group. Still, no double-positive cells for nestin/caspase-3 and GFAP/caspase-3 were found after ketamine exposure. In addition, the neuronal differentiation of NSCs in the SVZ was markedly promoted by ketamine with an increased number of ß-tubulin III/BrdU double-positive cells and enhanced expression of ß-tubulin III. These effects of ketamine on the NSCs in the SVZ often lasted at least 1 week after ketamine anesthesia. CONCLUSION: In the present study, it was demonstrated that ketamine could alter neurogenesis by inhibiting the proliferation of NSCs, suppressing their differentiation into astrocytes and promoting the neuronal differentiation of the NSCs in the SVZ of neonatal rats during a critical period of their neurodevelopment.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ketamina/farmacologia , Ventrículos Laterais/metabolismo , Animais , Animais Recém-Nascidos , Anticorpos/imunologia , Apoptose/efeitos dos fármacos , Astrócitos/citologia , Caspase 3/imunologia , Caspase 3/metabolismo , Ventrículos Laterais/citologia , Masculino , Nestina/imunologia , Nestina/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Tubulina (Proteína)/imunologia , Tubulina (Proteína)/metabolismo
2.
Anesth Analg ; 120(6): 1361-8, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25695672

RESUMO

BACKGROUND: In this study, we investigated the effect of propofol, a commonly used IV anesthetic, on lipopolysaccharide (LPS)-induced inflammatory responses in astrocytes and explored the molecular mechanisms by which it occurs. METHODS: Astrocytes were stimulated with LPS (1.0 µg/mL) in the absence and presence of different concentrations of propofol. The expression of astrocyte marker glial fibrillary acidic protein (GFAP) in astrocytes was detected using immunofluorescence staining and Western blot analysis. The levels of interleukin (IL)-1ß, IL-6, and tumor necrosis factor-α were measured using an enzyme-linked immunosorbent assay. The mRNA level of Toll-like receptor 4 (TLR4) was determined by semiquantitative reverse transcriptase-polymerase chain reaction. The protein expressions of TLR4, myeloid differentiation factor 88 (MyD88), p- extracellular signal-regulated protein kinases (ERK)1/2, p-c-Jun N-terminal kinase, p-p38 mitogen-activated protein kinase (MAPK), p-I-κBα, I-κBα, and p-nuclear factor-κB (NF-κB)p65 were detected by Western blot. RESULTS: Our results show that after stimulation with LPS, the levels of IL-1ß, IL-6, and tumor necrosis factor-α and the expression of GFAP in astrocytes were up-regulated significantly. In addition, the expression of TLR4, MyD88, p-ERK1/2, p-c-Jun N-terminal kinase, p-p38 MAPK, and p-NF-κBp65 increased, whereas the expression of total I-κBα decreased upon stimulation with LPS. Propofol (10 µM) reduced the secretion of proinflammatory cytokines, inhibited the expressions of GFAP, TLR4, MyD88, p-ERK1/2, p-p38 MAPK, and p-NF-κBp65 in astrocytes challenged with LPS. CONCLUSIONS: In the present study, propofol 10 µM but not lower clinically relevant or higher supra-clinical concentrations attenuated LPS-induced astrocyte activation and subsequent inflammatory responses by inhibiting the TLR4/MyD88-dependent NF-κB, ERK1/2, and p38 MAPK pathways.


Assuntos
Anti-Inflamatórios/farmacologia , Astrócitos/efeitos dos fármacos , Lipopolissacarídeos/toxicidade , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Fármacos Neuroprotetores/farmacologia , Propofol/farmacologia , Receptor 4 Toll-Like/efeitos dos fármacos , Fator de Transcrição RelA/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Animais Recém-Nascidos , Astrócitos/enzimologia , Astrócitos/imunologia , Células Cultivadas , Citocinas/metabolismo , Citoproteção , Relação Dose-Resposta a Droga , Proteína Glial Fibrilar Ácida/metabolismo , Mediadores da Inflamação/metabolismo , Fosforilação , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
3.
Cell Physiol Biochem ; 34(5): 1792-801, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25427956

RESUMO

BACKGROUND/AIMS: Ketamine is a widely used anesthetic in obstetric and pediatric anesthesia. In the developing brain, the widespread neuron apoptosis triggered by ketamine has been demonstrated. However, little is known about its effect on neural stem cells (NSCs) function. This study aimed to investigate the effect of ketamine on proliferation of NSCs from neonatal rat hippocampus. METHODS: Neural stem cells were isolated from the hippocampus of Sprague-Dawley rats on postnatal day 3. In dose-response experiments, cultured neural stem cells (NSCs) were exposed to different concentrations of ketamine (0-1000 µM) for 24 hrs. The proliferative activity of NSCs was evaluated by 5-Bromo-2'-deoxyuridine (BrdU) incorporation assay. Apoptosis of neural stem cells were assessed using caspase-3 by western blot. The intracellular Ca(2+) concentration ([Ca(2+)]i) in NSCs was analyzed by flow cytometry. The activation of protein kinase C-α (PKCα) and the phosphorylation of extracellular signal-regulated kinases 1/2 (ERK1/2) were measured by western blot analysis. RESULTS: Clinical relevant concentration of ketamine (10, 20 and 50 µM) did not markedly alter the proliferation of NSCs from neonatal rat hippocampus in vitro. However, ketamine (200, 500, 800 and 1000µM) significantly inhibited the proliferation of NSCs and did not affect the expression of caspase-3. Meanwhile, ketamine (200, 500, 800 and 1000µM) also markedly decreased [Ca(2+)]i as well as suppressed PKCα activation and ERK1/2 phosphorylation in NSCs. A combination of subthreshold concentrations of ketamine (100 µM) and Ca(2+) channel blocker verapamil (2.5 µM), PKCα inhibitor chelerythrine (2.5 µM) or ERK1/2 kinase inhibitor PD98059 (5 µM) significantly produced suprathreshold effects on PKCα activation, ERK1/2 phosphorylation and NSC proliferation. CONCLUSION: Ketamine inhibited proliferation of NSCs from neonatal rat hippocampus in vitro. Suppressing Ca(2+)-PKCα-ERK1/2 signaling pathway may be involved in this inhibitory effect of ketamine on NSCs proliferation.


Assuntos
Proliferação de Células/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Ketamina/farmacologia , Células-Tronco Neurais/efeitos dos fármacos , Animais , Animais Recém-Nascidos/metabolismo , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Caspase 3/metabolismo , Células Cultivadas , Hipocampo/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Fosforilação/efeitos dos fármacos , Proteína Quinase C-alfa/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
4.
J Endocrinol ; 232(2): 165-174, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27895138

RESUMO

Urotensin II (U-II), a cyclic peptide originally isolated from the caudal neurosecretory system of fishes, can produce proinflammatory effects through its specific G protein-coupled receptor, GPR14. Neuropathic pain, a devastating disease, is related to excessive inflammation in the spinal dorsal horn. However, the relationship between U-II and neuropathic pain has not been reported. This study was designed to investigate the effect of U-II antagonist on neuropathic pain and to understand the associated mechanisms. We reported that U-II and its receptor GPR14 were persistently upregulated and activated in the dorsal horn of L4-6 spinal cord segments after chronic constriction injury (CCI) in rats. Intrathecal injection of SB657510, a specific antagonist against U-II, reversed CCI-induced thermal hyperalgesia and mechanical allodynia. Furthermore, we found that SB657510 reduced the expression of phosphorylated c-Jun N-terminal kinase (p-JNK) and nuclear factor-κB (NF-κB) p65 as well as subsequent secretion of interleukin-1ß (IL-1ß), IL-6 and tumor necrosis factor-α (TNF-α). It was also showed that both the JNK inhibitor SP600125 and the NF-κB inhibitor PDTC significantly attenuated thermal hyperalgesia and mechanical allodynia in CCI rats. Our present research showed that U-II receptor antagonist alleviated neuropathic pain possibly through the suppression of the JNK/NF-κB pathway in CCI rats, which will contribute to the better understanding of function of U-II and pathogenesis of neuropathic pain.


Assuntos
Hiperalgesia/tratamento farmacológico , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , NF-kappa B/antagonistas & inibidores , Neuralgia/tratamento farmacológico , Sulfonamidas/uso terapêutico , Urotensinas/antagonistas & inibidores , Animais , Citocinas/metabolismo , Hiperalgesia/metabolismo , Masculino , Neuralgia/metabolismo , Estimulação Física , Ratos , Ratos Sprague-Dawley , Sulfonamidas/farmacologia
5.
Medicine (Baltimore) ; 95(28): e4082, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27428192

RESUMO

BACKGROUND: Trial design neuroinflammation and postoperative pain after surgery are increasingly reported in association with postoperative cognitive dysfunction (POCD). Parecoxib, a selective cyclooxygenase (COX)-2 inhibitor, is used for postoperative analgesia for its potent anti-inflammatory and analgesic effects. This study aimed to evaluate parecoxib's effects on POCD in elderly patients undergoing total knee arthroplasty. METHODS: Around 134 elderly patients undergoing total knee arthroplasty were randomly divided into parecoxib (group P) and control (group C) groups, and treated with parecoxib sodium and saline, respectively, shortly after induction of general anesthesia and 12-h postsurgery, respectively. Perioperative plasma IL-1ß, IL-6, TNF-α, and C-reactive protein (CRP) 1evels were measured. Postoperative pain was assessed following surgery. Neuropsychological tests were performed before surgery, and 1 week and 3 months postoperation. RESULTS: POCD incidence in group P was significantly lower compared with that of group C at 1 week after surgery (16.7% vs 33.9%; P < 0.05); no significant difference was found between groups C and P at 3-month follow-up (9.7% vs 6.7%). Compared with group C values, visual analog pain scale (VAS) scores at 3, 6, and 12 hours after surgery were significantly lower in group P(P < 0.05). Plasma IL-1ß, IL-6, and TNF-α levels were lower in group P than in group C after the operation (P < 0.05). No significant difference in the plasma CRP level was found between groups P and C. CONCLUSIONS: Parecoxib sodium decreases POCD incidence after total knee arthroplasty in elderly patients and may explain how this drug suppresses inflammation and acute postoperative pain caused by surgical trauma.


Assuntos
Artroplastia do Joelho , Disfunção Cognitiva/prevenção & controle , Inibidores de Ciclo-Oxigenase 2/uso terapêutico , Isoxazóis/uso terapêutico , Dor Pós-Operatória/prevenção & controle , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/sangue , Método Duplo-Cego , Feminino , Humanos , Masculino , Testes Neuropsicológicos
6.
Reg Anesth Pain Med ; 39(2): 137-48, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24513955

RESUMO

BACKGROUND AND OBJECTIVES: Neuropathic pain is related to the sustained activation of neuroglial cells and the production of proinflammatory cytokines in the spinal dorsal horn. Ghrelin, the endogenous ligand for growth hormone secretagogue receptor 1a (GHSR-1a), has been shown to inhibit the activation of microglia and the release of proinflammatory cytokines. The purpose of this study was to investigate the role of ghrelin/GHSR-1a signaling in neuropathic pain and to understand the associated mechanisms. METHODS: A rat model of neuropathic pain was established by chronic constriction injury (CCI) of the sciatic nerve. Hyperalgesia and allodynia were evaluated by observing the mechanical withdrawal threshold and the thermal withdrawal latency. The expression levels of ghrelin and GHSR-1a were detected by semiquantitative reverse transcriptase-polymerase chain reaction and Western blot analysis. The levels of interleukin-1ß (IL-1ß), IL-6, and tumor necrosis factor-α were detected using an enzyme-linked immunosorbent assay. The expression levels of p-p38 mitogen-activated protein kinases (p38 MAPK) and nuclear factor-κB (NF-κB) p65 were determined by Western blot and immunohistochemistry analysis. RESULTS: Both ghrelin and GHSR-1a were expressed in the spinal dorsal horns of normal rats and were not significantly different from that of sham rats. However, rats in the CCI model group developed severe hyperalgesia and allodynia, as well as significantly downregulated expression of ghrelin and GHSR-1a. Compared with CCI model rats, intrathecal injection of ghrelin clearly delayed thermal hyperalgesia and mechanical allodynia at 3, 5, and 7 days after CCI; reduced the levels of IL-1ß, IL-6, and tumor necrosis factor-α; and inhibited the phosphorylation of p38 MAPK and the activation of NF-κBp65 in the spinal dorsal horn. In addition, the effect of ghrelin could be blocked by [D-Lys]-GHRP-6, a GHSR-1a antagonist. CONCLUSIONS: Our present study demonstrated that ghrelin alleviated neuropathic pain through a GHSR-1a-mediated suppression of the p38 MAPK/NF-κB pathway.


Assuntos
Grelina/administração & dosagem , NF-kappa B/antagonistas & inibidores , Neuralgia/metabolismo , Receptores de Grelina/fisiologia , Neuropatia Ciática/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Animais , Doença Crônica , Constrição Patológica/tratamento farmacológico , Constrição Patológica/metabolismo , Grelina/biossíntese , Injeções Espinhais , Masculino , NF-kappa B/fisiologia , Neuralgia/tratamento farmacológico , Ratos , Ratos Sprague-Dawley , Neuropatia Ciática/tratamento farmacológico , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA