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










Base de datos
Intervalo de año de publicación
1.
Cogn Neurodyn ; 17(6): 1541-1559, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37974577

RESUMEN

The thalamocortical system plays an important role in consciousness. How anesthesia modulates the thalamocortical interactions is not completely known.  We simultaneously recorded local field potentials(LFPs) in thalamic reticular nucleus(TRN) and ventroposteromedial thalamic nucleus(VPM), and electrocorticographic(ECoG) activities in frontal and occipital cortices in freely moving rats (n = 11). We analyzed the changes in thalamic and cortical local spectral power and connectivities, which were measured with phase-amplitude coupling (PAC), coherence and multivariate Granger causality, at the states of baseline, intravenous infusion of propofol 20, 40, 80 mg/kg/h and after recovery of righting reflex. We found that propofol-induced burst-suppression results in a synchronous decrease of spectral power in thalamus and cortex (p < 0.001 for all frequency bands). The cross-frequency PAC increased by propofol, characterized by gradually stronger 'trough-max' pattern in TRN and stronger 'peak-max' pattern in cortex. The cross-region PAC increased in the phase of TRN modulating the amplitude of cortex. The functional connectivity (FC) between TRN and cortex for α/ß bands also significantly increased (p < 0.040), with increased directional connectivity from TRN to cortex under propofol anesthesia. In contrast, the corticocortical FC significantly decreased (p < 0.047), with decreased directional connectivity from frontal cortex to occipital cortex. However, the thalamothalamic functional and directional connectivities remained largely unchanged by propofol anesthesia.  The spectral powers and connectivities are differentially modulated with the changes of propofol doses, suggesting the changes in neural dynamics in thalamocortical system could be used for distinguishing different vigilance levels caused by propofol. Supplementary Information: The online version contains supplementary material available at 10.1007/s11571-022-09912-0.

2.
Int Immunopharmacol ; 120: 110349, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37210913

RESUMEN

Methylene blue (MB) has anti-inflammatory properties, however, its underlying molecular mechanism remains elusive. This study aimed to investigate whether and how MB could attenuate lipopolysaccharide (LPS)-induced microglial activation, neuroinflammation, and neurobehavioral deficits. We measured the expression of pro-inflammatory factors and performed three neurobehavioral tests to assess the effect of MB on neuroinflammation and neurocognitive dysfunction in LPS-treated adult C57BL/6N male mice or LPS-stimulated microglia cells. In vitro and in vivo experiments were further performed to investigate the molecular mechanism underlying MB inhibition of neuroinflammation using various experimental methods, including western blot, RT-qPCR, immunofluorescence, seahorse measurement, positron emission tomography (PET) scan, and flow cytometry analyses. Our results demonstrated that microglial activation and M1 polarization were induced by LPS exposure, resulting in an inflammatory response and neuronal apoptosis. Furthermore, LPS induced metabolic reprogramming in microglial cells. However, MB treatment substantially inhibited LPS-induced elevated levels of pro-inflammatory factors and reversed metabolic activation in vivo, which eventually led to the resolution of neuroinflammation and neurobehavioral improvement. Mechanistically, MB specifically inhibited the LPS-induced overexpression of PHD3 in vitro and in vivo. The pharmacological and genetic manipulations unveiled that the Siah2/Morg1/PHD3 signaling pathway may mediate MB protection against LPS-induced neuroinflammation and neurotoxicity. Therefore MB inhibited PHD3-dependent neuroinflammation may via Siah2/Morg1/PHD3 pathway, and that PHD3 expressed in microglia may be a drug target for the treatment of neuroinflammation-related brain disorders.


Asunto(s)
Inflamación , Microglía , Ratones , Animales , Masculino , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Enfermedades Neuroinflamatorias , Azul de Metileno/farmacología , Azul de Metileno/uso terapéutico , Azul de Metileno/metabolismo , Ratones Endogámicos C57BL , Ratones Endogámicos , Ubiquitina-Proteína Ligasas/metabolismo
4.
Neuroscience ; 481: 47-59, 2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-34801658

RESUMEN

It is well known that neuroinflammation plays a key role in neurodegenerative diseases. Hypoxia-inducible factor (HIF) and its hydroxylases-Prolyl-4-hydroxyases (PHDs) have been found to modulate the inflammatory processes. Here, the effects of PHDs enzyme onlipopolysaccharide-induced neuroinflammation and neurocognitive deficits were investigated. BV2 microglia cells were stimulated by LPS (1 µg/ml) as neuroinflammation model in vitro. Dimethyloxalylglycine (DMOG, 100 µM) and PHD3-siRNA were used to suppress the expression of PHD3. In vivo, mice received consecutive intraperitoneal injection of LPS (500 µg/kg) for 7 days, and intraperitoneal injection of DMOG (100 mg/kg) was applied 1 h before LPS at the same days. Several neurobehavioral tests (Open field, Novel object recognition and Morris water maze) were used to measure cognitive function. RT-qPCR and Western blotting were used to investigate the expression of inflammatory cytokines, HIF-PHDs protein. Metabolic reprogramming was measured by seahorse method. The results revealed that LPS induced neuroinflammation and PHD3 expression in vivo and vitro. DMOG and PHD3knockout decreased expression of inflammatory cytokines and improved the metabolic reprogramming caused by LPS treatment. Furthermore, pretreatment of DMOG reversed learning and memory deficits in systemic LPS-exposed mice through anti-neuroinflammation, which is independent of DMOG angiogenesis. These findings suggested that PHD3 may mediate LPS-induced microglial activation and neuroinflammation-associated neurobehavioral deficits.


Asunto(s)
Lipopolisacáridos , Enfermedades Neuroinflamatorias , Animales , Inflamación/inducido químicamente , Inflamación/metabolismo , Lipopolisacáridos/metabolismo , Lipopolisacáridos/toxicidad , Ratones , Microglía/metabolismo , Oxigenasas de Función Mixta/efectos adversos , Oxigenasas de Función Mixta/metabolismo
5.
ACS Chem Neurosci ; 10(5): 2332-2344, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30741526

RESUMEN

Cdh1 is a regulatory subunit of the anaphase promoting complex/cyclosome (APC/C), known to be involved in regulating neuronal survival. The role of Cdh1 in volatile anesthetics-induced neuronal apoptosis in the developing brain is unknown. In this study, we used postnatal day 7 (P7) and day 21 (P21) mice exposed to 2.3% sevoflurane for 6 h to investigate at which age and duration of exposure sevoflurane affects the expression of Cdh1 and glycolytic enzyme 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) and that of the pentose phosphate pathway (PPP) enzyme, glucose-6-phosphate dehydrogenase (G6PD). Furthermore, we tested whether the cyclin-dependent kinases (cdks) inhibitor roscovatine could counteract the effects caused by exposure to sevoflurane. Finally, we applied the glycolysis inhibitor 3-(3-pyridinyl)-1-(4-pyridinyl)-2-propen-1-one (3-PO), G6PD inhibitor dehydroepiandrosterone (DHEA), and exogenous reduced glutathione to examine the contribution of the glycolysis pathway and PPP to sevoflurane-induced neuroapoptosis. We found that prolonged sevoflurane anesthesia significantly reduces the Cdh1 level in P7 mice compared to in the P21 ones; moreover, the decrease in Cdh1 level results in a switch in glucose metabolism from the PPP to neuronal glycolysis. This leads to an imbalance between reactive oxygen species production and reduced glutathione level in the developing brain, which is more susceptible to oxidative stress. As a result, sevoflurane induces neuroapoptosis through Cdh1-mediated glucose metabolism reprogramming. Our study demonstrates a critical role of Cdh1 in sevoflurane-induced neuroapoptosis by shifting PPP to the glycolytic pathway in the developing brain. These findings suggest that Cdh1 may be a novel target for preventing volatile anesthetics-induced neurotoxicity and memory impairment.


Asunto(s)
Anestésicos por Inhalación/farmacología , Apoptosis/efectos de los fármacos , Encéfalo/metabolismo , Cadherinas/metabolismo , Vía de Pentosa Fosfato/efectos de los fármacos , Sevoflurano/farmacología , Animales , Biomarcadores/metabolismo , Glucemia/metabolismo , Encéfalo/crecimiento & desarrollo , Metabolismo Energético/efectos de los fármacos , Femenino , Glucólisis/efectos de los fármacos , Masculino , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo
6.
Brain Res Bull ; 146: 28-39, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30553844

RESUMEN

BACKGROUND: ER stress is involved in sevoflurane-induced neurotoxicity. Protein tyrosine phosphatase 1B (PTP1B) resided in the ER membrane is known to regulate ER stress. However, the role of PTP1B in sevoflurane-induced neurotoxicity is unknown. METHOD: Seven-day-old mice treated with 2.3% sevoflurane for 6 h as animal model. The hippocampal tissues were harvested following sevoflurane exposure for evaluation of ER stress markers with Western blot, ER morphology with transmission electron microscopy and density of dendrite spine with Golgi staining. In another subset of mice, neurocognitive function was assessed 4 weeks after anesthesia using Morris water maze test. We also examined the effects of PTP1B or PERK inhibitor on sevoflurane-induced neurodegeneration in the hippocampus. RESULT: The results showed inhibition of PTP1B significantly ameliorated sevoflurane-induced (1) ultrastructural ER alternations and ER stress activation as indicated by upregulation of PERK and eIF2α phosphorylation; (2) increase in cleaved caspase-3 expression; (3) elevated expressions of proinflammatory NF-κB and TNFα;(4) decreases in expression of synaptophysin, Arc and BDNF-TrkB proteins as well as loss of dendrite spine in the hippocampus; and (5) impairment in neurobehavioural performance at adolescence. Similarly, ER stress inhibitor is also found to downregulate eIF2α phosphorylation and neuroinflammation, and increase expressions of synaptic proteins and BDNF-TrkB signaling proteins. CONCLUSIONS: Our study shows PTP1B inhibition mitigates sevoflurane-induced neurodegeneration maybe mediated by ER stress in developing brain, and eventually improves cognitive function. This suggests PTP1B inhibition could represent a promising strategy to prevent sevoflurane-induced neurotoxicity.


Asunto(s)
Estrés del Retículo Endoplásmico/fisiología , Síndromes de Neurotoxicidad/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Sevoflurano/toxicidad , Animales , Encéfalo/embriología , Encéfalo/metabolismo , Caspasa 3/metabolismo , Espinas Dendríticas/metabolismo , Retículo Endoplásmico/metabolismo , Factor 2 Eucariótico de Iniciación/metabolismo , Femenino , Hipocampo/embriología , Hipocampo/metabolismo , Masculino , Ratones , Ratones Endogámicos , Modelos Animales , FN-kappa B/metabolismo , Síndromes de Neurotoxicidad/etiología , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Sevoflurano/farmacología , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , eIF-2 Quinasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA