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.
CNS Neurosci Ther ; 29(10): 2972-2985, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37287422

RESUMO

AIMS: Mid-gestational sevoflurane exposure may induce notable long-term neurocognitive impairment in offspring. This study was designed to investigate the role and potential mechanism of ferroptosis in developmental neurotoxicity induced by sevoflurane in the second trimester. METHODS: Pregnant rats on day 13 of gestation (G13) were treated with or without 3.0% sevoflurane, Ferrostatin-1 (Fer-1), PD146176, or Ku55933 on three consecutive days. Mitochondrial morphology, ferroptosis-relative proteins, malondialdehyde (MDA) levels, total iron content, and glutathione peroxidase 4 (GPX4) activities were measured. Hippocampal neuronal development in offspring was also examined. Subsequently, 15-lipoxygenase 2 (15LO2)-phosphatidylethanolamine binding protein 1 (PEBP1) interaction and expression of Ataxia telangiectasia mutated (ATM) and its downstream proteins were also detected. Furthermore, Morris water maze (MWM) and Nissl's staining were applied to estimate the long-term neurotoxic effects of sevoflurane. RESULTS: Ferroptosis mitochondria were observed after maternal sevoflurane exposures. Sevoflurane elevated MDA and iron levels while inhibiting GPX4 activity, and resultant long-term learning and memory dysfunction, which were alleviated by Fer-1, PD146176, and Ku55933. Sevoflurane could enhance 15LO2-PEBP1 interaction and activate ATM and its downstream P53/SAT1 pathway, which might be attributed to excessive p-ATM nuclear translocation. CONCLUSION: This study proposes that 15LO2-mediated ferroptosis might contribute to neurotoxicity induced by maternal sevoflurane anesthesia during the mid-trimester in the offspring and its mechanism may be ascribed to hyperactivation of ATM and enhancement of 15LO2-PEBP1 interaction, indicating a potential therapeutic target for ameliorating sevoflurane-induced neurotoxicity.


Assuntos
Ferroptose , Gravidez , Feminino , Ratos , Animais , Sevoflurano/toxicidade , Ratos Sprague-Dawley , Encéfalo/metabolismo , Ferro/metabolismo
2.
J Clin Anesth ; 86: 111075, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36796214

RESUMO

BACKGROUND: Pneumoperitoneum is a risk factor for perioperative atelectasis in infants. This research aimed to investigate whether lung recruitment manoeuvres guided by ultrasound are more effective for young infants (<3 months) during laparoscopy under general anaesthesia. METHODS: Young infants (<3 months) undergoing general anaesthesia during laparoscopic surgery (>2 h) were randomised to either conventional lung recruitment (control group) or ultrasound-guided lung recruitment (ultrasound group) once per hour. Mechanical ventilation was started with a tidal volume of 8 mL·kg-1, positive end-expiratory pressure of 6 cm H2O and 40% inspired oxygen fraction. Lung ultrasound (LUS) was performed four times (T1 was performed 5 min after intubation and before pneumoperitoneum set, T2 was performed after pneumoperitoneum, T3 was performed 1 min after surgery, and T4 was performed before being discharged from post-anaesthesia care unit [PACU]) in each infant. The primary outcome was the incidence of significant atelectasis at T3 and T4 (defined by LUS consolidation score ≥ 2 in any region). RESULTS: 62 babies were enrolled in the experiment and 60 infants were included in the analysis. Before the recruitment, atelectasis was similar between infants randomised to the control or ultrasound group at T1 (83.3% vs 80.0%; P = 0.500) and T2 (83,3% vs 76.7%; P = 0.519). The incidence of atelectasis at T3 and T4 were lower in the ultrasound group (26.7% and 33.3%), compared with infants randomised to conventional lung recruitment (66.7% and 70%) (P = 0.002; P = 0.004; respectively). CONCLUSIONS: Ultrasound-guided alveolar recruitment reduced the perioperative incidence of atelectasis in infants <3 months during laparoscopy under general anaesthesia.


Assuntos
Laparoscopia , Pneumoperitônio , Atelectasia Pulmonar , Lactente , Humanos , Pneumoperitônio/complicações , Atelectasia Pulmonar/diagnóstico por imagem , Atelectasia Pulmonar/etiologia , Atelectasia Pulmonar/prevenção & controle , Pulmão/diagnóstico por imagem , Laparoscopia/efeitos adversos , Ultrassonografia de Intervenção
3.
CNS Neurosci Ther ; 28(12): 2268-2280, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36184790

RESUMO

AIMS: Hypoxic-ischemic brain injury (HIBI) often results in cognitive impairments. Herein, we investigated the roles of ferroptosis in HIBI and the underlying signaling pathways. METHODS: Ferrostatin-1 (Fer-1) or resveratrol (Res) treatments were administered intracerebroventricularly 30 min before HIBI in 7-day-old rats. Glutathione peroxidase 4 (GPx4) expression, malondialdehyde (MDA) concentration, iron content, mitochondrial morphology, and the expression of silent information regulator factor 2-related enzyme 1 (SIRT1) and nuclear factor erythroid-2-related factor 2 (Nrf2) were measured after HIBI. Additionally, the weight ratio of left/right hemisphere, brain morphology, Nissl staining, and the Morris water maze test were conducted to estimate brain damage. RESULTS: At 24-h post-HIBI, GPx4 expression was decreased, and MDA concentration and iron content were increased in the hippocampus. HIBI led to mitochondrial atrophy, brain atrophy/damage, and resultant learning and memory impairments, which were alleviated by Fer-1-mediated inhibition of ferroptosis. Furthermore, Res-mediated SIRT1 upregulation increased Nrf2 and GPx4 expression, thereby attenuating ferroptosis, reducing brain atrophy/damage, and improving learning and memory abilities. CONCLUSION: The results demonstrated that during HIBI, ferroptosis occurs via the SIRT1/Nrf2/GPx4 signaling pathway, suggesting it as a potential therapeutic target for inhibiting ferroptosis and ameliorating HIBI-induced cognitive impairments.


Assuntos
Lesões Encefálicas , Ferroptose , Hipóxia-Isquemia Encefálica , Animais , Ratos , Fator 2 Relacionado a NF-E2/metabolismo , Sirtuína 1/metabolismo , Animais Recém-Nascidos , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Transdução de Sinais , Atrofia , Ferro
4.
Cell Mol Neurobiol ; 42(6): 1777-1786, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33606156

RESUMO

With the increase in fetal surgeries, the effect of maternal anesthesia on progeny has attracted much attention. Our previous studies have demonstrated that 3.5% sevoflurane maternal exposure resulted in over-activated autophagy and cognitive impairment in the offspring. The autophagy activation resulted in increased apoptosis and decreased proliferation. However, the effects of sevoflurane on neural stem cell (NSC) differentiation is unclear. There is evidence that autophagy might participate in anesthesia-induced NSC differentiation. Firstly, we examined the effects of sevoflurane on NSC differentiation and explored possible mechanisms. Then, we investigated whether autophagy was related to differentiation. On gestational day 14 (G14), rats were exposed to 2% or 3.5% sevoflurane for 2 h, then markers of neurons and astrocytes, and the FOXO3 expression was measured in fetal brains 48 h later. The differentiation of NSCs was detected after autophagy inhibition by 3-MA. Changes in NSC differentiation, autophagy level, and FOXO3 were examined after administration of lithium chloride. After 3.5% sevoflurane exposure, the expressions of ß-Tubulin III, NeuN, SYP, GFAP and FOXO3 increased. Autophagy inhibition alleviates improper NSC differentiation. Lithium chloride attenuated FOXO3 and autophagy activation, ameliorated NSC differentiation and the decline of Nestin expression. Our results demonstrated that maternal exposure to 3.5% sevoflurane for 2 h during the mid-trimester induced NSC differentiation in the fetal brain through the activation of FOXO3. Autophagy inhibitor or lithium chloride reversed the improper differentiation of NSCs.


Assuntos
Cloreto de Lítio , Células-Tronco Neurais , Animais , Diferenciação Celular , Feminino , Cloreto de Lítio/metabolismo , Cloreto de Lítio/farmacologia , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Ratos , Sevoflurano/metabolismo , Sevoflurano/farmacologia
5.
Neurochem Int ; 150: 105198, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34601014

RESUMO

Post-conditioning with sevoflurane, a volatile anesthetic, has been proved to be neuroprotective against hypoxic-ischemic brain injury (HIBI). Our previous research showed that autophagy is over-activated in a neonatal HIBI rat model, and inhibition of autophagy confers neuroprotection. There is increasing recognition that autophagy can be stimulated by activating endoplasmic reticulum (ER) stress. Herein, we purposed to explore: i) the association of ER stress with autophagy in the setting of neonatal HIBI; and ii) the possible roles of ER stress-triggered autophagy, as well as IRE1 signaling in the neuroprotection of sevoflurane post-conditioning against neonatal HIBI. Seven-day-old rats underwent ligation of the left common artery, and a subsequent 2 h hypoxia (8% O2/92% N2). The association of ER stress with autophagy was examined by ER stress inducer (tunicamycin), 4-PBA (ER stress inhibitor), or 3-MA (autophagy inhibitor). Rats in the sevoflurane post-conditioning groups were treated with 2.4% sevoflurane for 30 min after HIBI stimulation. The roles of ER stress-mediated autophagy, as well as the IRE1-JNK-beclin1 signaling cascade in the neuroprotection afforded by sevoflurane were explored by ER stress inducer (tunicamycin) and the IRE1 inhibitor (STF-083010). HIBI over-activated ER stress and autophagy in neonatal rats. HIBI-induced autophagy was significantly aggravated by tunicamycin but blocked by 4-PBA; however, HIBI-induced ER stress was not affected by 3-MA. Sevoflurane post-conditioning significantly alleviated ER stress, autophagy, cell apoptosis, and cognitive impairments, which were remarkably abolished by tunicamycin. Also, tunicamycin blocked sevoflurane-induced downregulation of IRE1-JNK-beclin1 signaling pathway. Whereas, IRE1 inhibitor could reverse the effects of tunicamycin. ER stress contributes to autophagy induced by HIBI. Furthermore, sevoflurane post-conditioning significantly protects against HIBI in neonatal rats by inhibiting ER stress-mediated autophagy via IRE1-JNK-beclin1 signaling cascade.


Assuntos
Autofagia/fisiologia , Estresse do Retículo Endoplasmático/fisiologia , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Pós-Condicionamento Isquêmico/métodos , Proteínas de Membrana/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Sevoflurano/administração & dosagem , Animais , Animais Recém-Nascidos , Autofagia/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Hipóxia-Isquemia Encefálica/metabolismo , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Proteínas de Membrana/metabolismo , Inibidores da Agregação Plaquetária/administração & dosagem , Proteínas Serina-Treonina Quinases/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
6.
Neurotox Res ; 39(5): 1524-1542, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34224102

RESUMO

Sevoflurane postconditioning (SPC) has been widely reported to attenuate brain injury after hypoxia-ischemia encephalopathy (HIE) by inhibiting neural necrosis and autophagy. Moreover, recent reports revealed that sevoflurane facilitated hippocampal reconstruction via regulating migration. Yet, it remains unclear whether the promotion of neural migration by SPC repairs the hippocampal injury after HIE. Here, we hypothesize that SPC exerts a neuroprotective effect by ameliorating neuronal migration disorder after HIE and regulating Reelin expression. Furthermore, the downstream Reelin/Dab1 pathway may be involved. The classical Rice-Vannucci model of hypoxia-ischemia was performed on postnatal day 7 rat pups, which was followed by SPC at 1 minimum alveolar concentration (MAC 2.5%) for 30 min. Piceatannol, causing Reelin aggregation in vivo, was used to detect whether Reelin/Dab1 was involved in the neuroprotection effect of SPC. Hippocampal-dependent learning ability tests were conducted to assess the long-term effects on locomotor activity and spatial learning ability. Our findings suggest that hypoxia-ischemia injury inhibited neurons migrated outward from the basal zone of dentate gyrus, disrupted cytoarchitecture of the dentate gyrus (DG), and led to long-term cognition deficits. However, SPC could relieve the restricted hippocampal neurons and repair the hippocampal-dependent memory function damaged after HIE by attenuating the overactivation of the Reelin/Dab1 pathway. These results demonstrate that SPC plays a pivotal role in ameliorating neuronal migration disorder and maintaining normal cytoarchitecture of the DG via inhibiting overactivated Reelin expression. This process may involve overactivated Reelin/Dab1 signaling pathway and spatial learning ability by regulating the Reelin expression which may associate with its neuroprotection.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Cognição/efeitos dos fármacos , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Malformações do Desenvolvimento Cortical do Grupo II/tratamento farmacológico , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteína Reelina/antagonistas & inibidores , Sevoflurano/administração & dosagem , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Animais , Animais Recém-Nascidos , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Cognição/fisiologia , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/patologia , Pós-Condicionamento Isquêmico/métodos , Masculino , Malformações do Desenvolvimento Cortical do Grupo II/metabolismo , Malformações do Desenvolvimento Cortical do Grupo II/patologia , Proteínas do Tecido Nervoso/biossíntese , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/administração & dosagem , Inibidores da Agregação Plaquetária/administração & dosagem , Ratos , Ratos Sprague-Dawley , Proteína Reelina/biossíntese , Fatores de Tempo
7.
Life Sci ; 270: 118980, 2021 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-33428879

RESUMO

BACKGROUND: Hypoxic-ischemic brain injury (HIBI) is a major cause of mortality in neonates and can cause long-term neurological sequelae. Excessive autophagy caused by HI may cause neuronal death. Dexmedetomidine was reported neuroprotective against HIBI. Therefore, in the present study, the autophagy-related mechanisms underlying the protective effects of dexmedetomidine against cerebral HI in neonatal rats were investigated. METHODS: In the present study, the expression of autophagy-related proteins microtubule-associated protein 1 light chain 3 (LC3) B-II and Beclin1, neuronal and microglia autophagy levels, the myelin basic protein (MBP) expression, long-term neuronal density ratio, and long-term behavioral prognosis in HIBI model were investigated by ligating the left common carotid artery in neonatal rats, followed by 2-h hypoxia. RESULTS: Dexmedetomidine inhibited the overactivated autophagy of hippocampal neurons and microglia after HI. In addition, dexmedetomidine inhibited neuronal density decrease and axon demyelination after HI-induced overactivated autophagy. Lastly, dexmedetomidine improved the long-term neurological prognosis and was reversed by the autophagy agonist rapamycin. CONCLUSION: The protective effects of dexmedetomidine on HI neonatal rats were evidenced by inhibition of excessive autophagy of neurons and microglia, thereby reducing the decline of long-term neuronal density and axon demyelination as well as improving long-term learning cognitive function.


Assuntos
Dexmedetomidina/farmacologia , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Pós-Condicionamento Isquêmico/métodos , Animais , Animais Recém-Nascidos , Autofagia/efeitos dos fármacos , Encéfalo/metabolismo , Morte Celular/efeitos dos fármacos , Dexmedetomidina/metabolismo , Feminino , Hipocampo/metabolismo , Hipóxia/metabolismo , Isquemia/metabolismo , Masculino , Microglia/metabolismo , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Sprague-Dawley
8.
Cell Mol Neurobiol ; 41(8): 1801-1816, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32880098

RESUMO

Microglia/macrophages have been identified to be highly polarized after ischemia. Interestingly, the polarization of these microglia/macrophages varies immensely under differing disease conditions. Post-conditioning using sevoflurane, a volatile anesthetic, could provide long-term neuroprotection to neonatal rats after hypoxic-ischemic brain injury (HIBI). Thus, the current study aimed at investigating the effects of sevoflurane post-conditioning (SPC) on microglia/macrophage polarization after HIBI induction in neonatal rats. Additionally, we aimed at identifying the underpinning mechanisms specifically related to autophagy and lysosomal protease enzyme, cathepsin B. To develop a HIBI model, 7-day-old Sprague-Dawley rats underwent left common carotid artery ligation followed by 2 h of hypoxia. The role of microglia/macrophages in the neuroprotection conferred by SPC was examined by left-side intra-cerebroventricular injection with adenovirus vector carrying catB-GFP or rapamycin. The number of interleukin (IL)-1ß+ cells, cathepsin B+ cells, light chain 3B positive (LC3B+) cells among ionized calcium binding adaptor molecule 1(Iba1+)cells to investigate microglia polarization, neuronal apoptosis to assess neuronal death in the acute phase were tested at 24 h after HIBI. Behavioral tests including suspension test, Morris water maze tests were performed to investigate the long-term effects of SPC, at 21 to 34 days post HIBI. Nissl staining and myelin basic protein (MBP) immunostaining to assess the long-term neuronal and myelin damage were performed at 34 days after HIBI. Based on the obtained results post HIBI, we observed the cells that were positive for IL-1ß, cathepsin B, and LC3B among Iba1 positive cell population in the hippocampus were significantly decreased after SPC treatment. SPC significantly attenuated the HIBI-induced increase in neuronal apoptosis, improved long-term cognitive function, and attenuated HI-induced decrease of Nissl-positive cells and MBP expression. However, these trends were reversed by injection of adenovirus vector carrying catB-GFP and rapamycin. SPC attenuated microglia polarization towards neurotoxic phenotypes, alleviates neuronal death and axon demyelination after HIBI in neonatal rats by regulating microglia autophagy and cathepsin B expression, and therefore provided long-term cognitive, learning and memory protection.


Assuntos
Doenças Desmielinizantes/terapia , Hipóxia-Isquemia Encefálica/terapia , Pós-Condicionamento Isquêmico/métodos , Macrófagos/efeitos dos fármacos , Microglia/efeitos dos fármacos , Sevoflurano/administração & dosagem , Animais , Animais Recém-Nascidos , Axônios/efeitos dos fármacos , Axônios/metabolismo , Encéfalo/irrigação sanguínea , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Doenças Desmielinizantes/metabolismo , Feminino , Hipóxia-Isquemia Encefálica/metabolismo , Macrófagos/metabolismo , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Microglia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Inibidores da Agregação Plaquetária/administração & dosagem , Ratos , Ratos Sprague-Dawley
9.
Neural Regen Res ; 16(6): 1052-1061, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33269750

RESUMO

Previous studies have demonstrated that sevoflurane postconditioning can provide neuroprotection after hypoxic-ischemic injury and improve learning and memory function in developing rodent brains. The classical Rice-Vannucci model was used to induce hypoxic-ischemic injury, and newborn (postnatal day 7) rats were treated with 2.4% sevoflurane for 30 minutes after hypoxic-ischemic injury. Our results showed that sevoflurane postconditioning significantly improved the learning and memory function of rats, decreased astrogliosis and glial scar formation, increased numbers of dendritic spines, and protected the histomorphology of the hippocampus. Mechanistically, sevoflurane postconditioning decreased expression of von Hippel-Lindau of hypoxia-inducible factor-1α and increased expression of DJ-1. Injection of 1.52 µg of the hypoxia-inducible factor-1α inhibitor YC-1 (Lificiguat) into the left lateral ventricle 30 minutes before hypoxic-ischemic injury reversed the neuroprotection induced by sevoflurane. This finding suggests that sevoflurane can effectively alleviate astrogliosis in the hippocampus and reduce learning and memory impairments caused by glial scar formation after hypoxic-ischemic injury. The underlying mechanism may be related to upregulated DJ-1 expression, reduced ubiquitination of hypoxia-inducible factor-1α, and stabilized hypoxia-inducible factor-1α expression. This study was approved by the Laboratory Animal Care Committee of China Medical University, China (approval No. 2016PS337K) on November 9, 2016.

10.
Biomed Pharmacother ; 131: 110733, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33152912

RESUMO

General anaesthesia may impose significant neurocognitive risks on the developing brain. As brain injury in preterm neonates has a particular predilection for cerebral white matter, we aimed to evaluate the effects of sevoflurane on oligodendrocyte maturation and myelination in a preterm-equivalent rat model. Two-day-old postnatal (P2) Sprague-Dawley rats were exposed to 3.3 % (approximately 1 minimum alveolar concentration [MAC]) or 4.9 % (approximately 1.5 MAC) sevoflurane for 2 h. Physiologic parameters were measured at the end of sevoflurane anaesthesia. Oligodendrocyte proliferation, maturation, and myelination were evaluated by immunofluorescence with specific markers at different time points. Open field test and Morris water maze tests were performed to access behavior changes from P29 to P36. Arterial blood gases values and blood glucose levels were within the normal physiologic range. As compared to control, 4.9 %, but not 3.3 % sevoflurane disturbed oligodendrocyte maturation at P14, resulting in hypomyelination and axonal damage in cerebral white matter at P28. Rats exposed to 4.9 %, but not 3.3 % sevoflurane showed decreased explorative activity and increased anxiety-like behaviour, as well as learning and memory impairments. Furthermore, 4.9 %, but not 3.3 % sevoflurane inhibited oligodendrocyte proliferation in the developing white matter of the rat brain at 12 h post-anaesthesia, with further evidence of widespread reactive astrogliosis. High concentration of sevoflurane (4.9 %) exposure in early postnatal rats may disrupt oligodendrocyte maturation and myelination. Our study has aimed a spotlight on the need for safe and rational use of analgesics in neonates, especially preterm infants.


Assuntos
Bainha de Mielina/efeitos dos fármacos , Oligodendroglia/efeitos dos fármacos , Sevoflurano/toxicidade , Substância Branca/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Astrócitos/fisiologia , Masculino , Teste do Labirinto Aquático de Morris , Proteína Básica da Mielina/análise , Bainha de Mielina/fisiologia , Oligodendroglia/fisiologia , Ratos , Ratos Sprague-Dawley , Substância Branca/fisiologia
11.
Drug Des Devel Ther ; 13: 1691-1706, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31190748

RESUMO

Background: When neonatal rats suffer hypoxic-ischemic brain injury (HIBI), autophagy is over-activated in the hippocampus, and inhibition of autophagy provides neuroprotection. The aim of this study was to investigate the possible roles of autophagy and Ezh2-regulated Pten/Akt/mTOR pathway in sevoflurane post-conditioning (SPC)-mediated neuroprotection against HIBI in neonatal rats. Methods: Seven-day-old Sprague-Dawley rats underwent left common artery ligation followed by 2 h hypoxia as described in the Rice-Vannucci model. The roles of autophagy and the Ezh2-regulated Pten/Akt/mTOR signaling pathway in the neuroprotection conferred by SPC were examined by left-side intracerebroventricular injection with the autophagy activator rapamycin and the Ezh2 inhibitor GSK126. Results: SPC was neuroprotective against HIBI through the inhibition of over-activated autophagy in the hippocampus as characterized by the rapamycin-induced reversal of neuronal density, neuronal morphology, cerebral morphology, and the expression of the autophagy markers, LC3B-II and Beclin1. SPC significantly increased the expression of Ezh2, H3K27me3, pAkt, and mTOR and decreased the expression of Pten induced by HI. The Ezh2 inhibitor, GSK126, significantly reversed the SPC-induced changes in expression of H3K27me3, Pten, pAkt, mTOR, LC3B-II, and Beclin1. Ezh2 inhibition also reversed SPC-mediated attenuation of neuronal loss and behavioral improvement in the Morris water maze. Conclusion: These results indicate that SPC inhibits excessive autophagy via the regulation of Pten/Akt/mTOR signaling by Ezh2 to confer neuroprotection against HIBI in neonatal rats.


Assuntos
Autofagia/efeitos dos fármacos , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Sevoflurano/farmacologia , Animais , Animais Recém-Nascidos , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Feminino , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/patologia , Indóis/farmacologia , Masculino , Piridonas/farmacologia , Ratos , Ratos Sprague-Dawley
12.
Drug Des Devel Ther ; 13: 4439-4449, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-32099322

RESUMO

BACKGROUND: Dexmedetomidine (Dex) is a highly selective α2-adrenoceptor agonist used as an off-label medication for pediatric sedation and analgesia. Recently, Dex was reported to exhibit neuroprotective efficacy in several brain injury models. Here we investigate whether neonatal Dex administration promotes hippocampal neurogenesis and enhances hippocampus-dependent spatial learning and memory under physiological conditions. METHODS: Postnatal day 7 (P7) pups were administered saline (vehicle control) or Dex (10, 20, or 40 µg/kg) by intraperitoneal injection. Neurogenesis and astrogenesis were examined in brain slices by BrdU immunostaining on P8 and changes in the expression levels of GDNF, NCAM, CREB, PSD95, and GAP43 were assessed by Western blotting on P35, respectively. Open field and Morris water maze (MWM) tests were conducted from P28 to P36 in order to assess effects on general motor activity and spatial learning, respectively. RESULTS: Dexmedetomidine at 20 µg/kg significantly enhanced neurogenesis and astrogenesis in hippocampus and upregulated GDNF, NCAM, CREB, PSD95, and GAP43 compared to vehicle and other Dex doses. Moreover, 20 µg/kg Dex-injected rats showed no changes in motor or anxiety-like behavior but performed better in the MWM test compared to all other groups. CONCLUSION: Neonatal injection of Dex (20 µg/kg) enhances spatial learning and memory in rat pups, potentially by promoting hippocampal neurogenesis and synaptic plasticity via activation of GDNF/NCAM/CREB signaling.


Assuntos
Dexmedetomidina/farmacologia , Hipocampo/efeitos dos fármacos , Hipnóticos e Sedativos/farmacologia , Aprendizagem em Labirinto/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Aprendizagem Espacial/efeitos dos fármacos , Animais , Dexmedetomidina/administração & dosagem , Feminino , Hipnóticos e Sedativos/administração & dosagem , Injeções Intraperitoneais , Masculino , Ratos , Ratos Sprague-Dawley
13.
Materials (Basel) ; 11(1)2017 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-29283411

RESUMO

The CrS/NbC Co-based self-lubricating composite coatings were successfully fabricated on Cr12MoV steel surface by laser clad Stellite 6, WS2, and NbC mixed powders. The phase composition, microstructure, and tribological properties of the coatings ware investigated by means of X-ray diffraction (XRD), scanning electron microscopy (SEM), and energy dispersive spectrometer (EDS), as well as dry sliding wear testing. Based on the experimental results, it was found reactions between WS2 and Co-based alloy powder had occurred, which generated solid-lubricant phase CrS, and NbC play a key role in improving CrS nuclear and refining microstructure of Co-based composite coating during laser cladding processing. The coatings were mainly composed of γ-Co, CrS, NbC, Cr23C6, and CoCx. Due to the distribution of the relatively hard phase of NbC and the solid lubricating phase CrS, the coatings had better wear resistance. Moreover, the suitable balance of CrS and NbC was favorable for further decreasing the friction and improving the stability of the contact surfaces between the WC ball and the coatings. The microhardness, friction coefficient, and wear rate of the coating 4 (Clad powders composed of 60 wt % Stellite 6, 30 wt % NbC and 10 wt % WS2) were 587.3 HV0.5, 0.426, and 5.61 × 10-5 mm³/N·m, respectively.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...