Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sleep ; 46(11)2023 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-36827092

RESUMEN

STUDY OBJECTIVES: This study verified that sleep deprivation before and after skin/muscle incision and retraction (SMIR) surgery increased the risk of chronic pain and investigated the underlying roles of microglial voltage-dependent anion channel 1 (VDAC1) signaling. METHODS: Adult mice received 6 hours of total sleep deprivation from 1 day prior to SMIR until the third day after surgery. Mechanical and heat-evoked pain was assessed before and within 21 days after surgery. Microglial activation and changes in VDAC1 expression and oligomerization were measured. Minocycline was injected to observe the effects of inhibiting microglial activation on pain maintenance. The VDAC1 inhibitor 4,4'-diisothiocyanostilbene-2,2'-disulfonic acid (DIDS) and oligomerization inhibitor VBIT-4 were used to determine the roles of VDAC1 signaling on microglial adenosine 5' triphosphate (ATP) release, inflammation (IL-1ß and CCL2), and chronicity of pain. RESULTS: Sleep deprivation significantly increased the pain duration after SMIR surgery, activated microglia, and enhanced VDAC1 signaling in the spinal cord. Minocycline inhibited microglial activation and alleviated sleep deprivation-induced pain maintenance. Lipopolysaccharide (LPS)-induced microglial activation was accompanied by increased VDAC1 expression and oligomerization, and more VDAC1 was observed on the cell membrane surface compared with control. DIDS and VBIT-4 rescued LPS-induced microglial ATP release and IL-1ß and CCL2 expression. DIDS and VBIT-4 reversed sleep loss-induced microglial activation and pain chronicity in mice, similar to the effects of minocycline. No synergistic effects were found for minocycline plus VBIT-4 or DIDS. CONCLUSIONS: Perioperative sleep deprivation activated spinal microglia and increases the risk of chronic postsurgical pain in mice. VDAC1 signaling regulates microglial activation-related ATP release, inflammation, and chronicity of pain.


Asunto(s)
Microglía , Privación de Sueño , Ratones , Animales , Microglía/metabolismo , Privación de Sueño/complicaciones , Privación de Sueño/metabolismo , Canal Aniónico 1 Dependiente del Voltaje/metabolismo , Minociclina/farmacología , Minociclina/metabolismo , Ácido 4,4'-Diisotiocianostilbeno-2,2'-Disulfónico/metabolismo , Lipopolisacáridos/metabolismo , Dolor Postoperatorio , Inflamación/metabolismo , Adenosina Trifosfato
2.
Sci Rep ; 6: 19869, 2016 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-26813160

RESUMEN

FGF10 is a member of fibroblast growth factors (FGFs). We previously showed that FGF10 protects neuron against oxygen-glucose deprivation injury in vitro; however, the effect of FGF10 in ischemic stroke in vivo is unknown. In the present study, we showed that FGF10 was mainly expressed in neurons but not astrocytes, and detected FGF10 in mouse cerebrospinal fluid. The FGF10 levels in neurons culture medium and cell lysate were much higher than those in astrocytes. FGF10 expression in brain tissue and FGF10 level in CSF were increased in mouse middle cerebral artery occlusion (MCAO) model. Administration of FGF10 into lateral cerebroventricle not only decreased MCAO-induced brain infarct volume and neurological deficit, but also reduced the number of TUNEL-positive cells and activities of Caspases. Moreover, FGF10 treatment depressed the triggered inflammatory factors (TNF-α and IL-6) and NF-κB signaling pathway, and increased phosphorylation of PI3K/Akt signaling pathway. Blockade of PI3K/Akt signaling pathway by wortmannin and Akt1/2-kinase inhibitor, partly compromised the neuroprotection of FGF10. However, blockade of PI3K/Akt signaling pathway did not impair the anti-inflammation action of FGF10. Collectively, our results demonstrate that neuron-derived FGF10 ameliorates cerebral ischemia injury via inhibiting NF-κB-dependent neuroinflammation and activating PI3K/Akt survival signaling pathway in mice.


Asunto(s)
Isquemia Encefálica/metabolismo , Factor 10 de Crecimiento de Fibroblastos/metabolismo , FN-kappa B/metabolismo , Neuronas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Daño por Reperfusión/metabolismo , Transducción de Señal , Animales , Apoptosis/efectos de los fármacos , Isquemia Encefálica/patología , Modelos Animales de Enfermedad , Factor 10 de Crecimiento de Fibroblastos/farmacología , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Modelos Biológicos , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Daño por Reperfusión/patología , Transducción de Señal/efectos de los fármacos
3.
J Arthroplasty ; 30(8): 1359-63, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25769742

RESUMEN

This study aimed to investigate the influence of dexmedetomidine (DEX) on the tourniquet related responses in hypertension patients receiving unilateral knee arthroplasty (UKA) under general anesthesia. Results showed that the incidence of tourniquet induced hypertension (TIH), hemodynamics, MAC and EtSEV in DEX group were significantly lower than those in control group, regardless of hypertension. However, significant differences in TIH, hemodynamics, minimum alveolar concentration (MAC) and end-tidal sevoflurane (EtSEV) were not observed between hypertension patients and non-hypertension patients in both control group and DEX group. Moreover, oxygen index (OI) and respiratory index (RI) remained unchanged after deflation and DEX failed to affect OI and RI within 30 min after deflation, regardless of hypertension. Taken together, DEX may significantly improve the hemodynamics, which is independent of pre-existing hypertension.


Asunto(s)
Anestesia General/métodos , Artroplastia de Reemplazo de Rodilla/métodos , Dexmedetomidina/uso terapéutico , Hipertensión/complicaciones , Torniquetes , Administración Intravenosa , Anciano , Analgésicos no Narcóticos/uso terapéutico , Antihipertensivos , Presión Sanguínea/efectos de los fármacos , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Hemodinámica , Humanos , Incidencia , Masculino , Éteres Metílicos/administración & dosificación , Persona de Mediana Edad , Oxígeno/química , Pruebas de Función Respiratoria , Frecuencia Respiratoria , Sevoflurano , Factores de Tiempo
4.
CNS Neurosci Ther ; 20(9): 851-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24948006

RESUMEN

BACKGROUND: Propofol is a short-acting, intravenous general anesthetic that is widely used in clinical practice for short procedures; however, it causes depressed cognitive function for several hours thereafter. (R)-alpha-methylhistamine (RAMH), a selective histamine H3 receptor agonist, can enhance memory retention and attenuates memory impairment in rats. In this study, we investigated whether RAMH could rescue propofol-induced memory deficits and the underlying mechanisms partaking in this process. METHODS: In the modified Morris water maze (MWM) test, rats were randomized into the following groups: control, propofol (25 mg/kg, i.p., 30 min before training), RAMH (10 mg/kg, i.p., 60 min before training), and propofol plus RAMH. All randomized rats were subjected to 2 days of training, and a probe test was conducted on day 3. Field excitatory postsynaptic potentials were recorded from CA1 neurons in rat hippocampal slices, and long-term potentiation (LTP) was induced by either theta-burst stimulation (TBS) or high-frequency tetanic stimulation (HFS). Spontaneous and miniature inhibitory (sIPSCs, mIPSCs) or excitatory (sEPSCs, mEPSCs) postsynaptic currents were recorded from CA1 pyramidal neurons by whole-cell patch clamp. RESULTS: In the MWM task, propofol injection significantly impaired spatial memory retention. Pretreatment with RAMH reversed propofol-induced memory retention. In hippocampal CA1 slices, propofol perfusion markedly inhibited TBS- but not HFS-induced LTP. Co-perfusion of RAMH reversed the inhibitory effect of propofol on TBS-induced LTP reduction. Furthermore, in hippocampal CA1 pyramidal neurons, RAMH significantly suppressed the frequency but not the amplitude of sIPSCs and mIPSCs and had little effects on both the frequency and amplitude of sEPSCs and mEPSCs. CONCLUSIONS: Our results suggest that RAMH, by inhibiting presynaptic GABAergic neurotransmission, suppresses inhibitory neurotransmission in hippocampal CA1 pyramidal neurons, which in turn reverses inhibition of CA1 LTP and the spatial memory deficits induced by propofol in rats.


Asunto(s)
Amnesia/tratamiento farmacológico , Región CA1 Hipocampal/citología , Agonistas de los Receptores Histamínicos/uso terapéutico , Metilhistaminas/uso terapéutico , Células Piramidales/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Amnesia/inducido químicamente , Amnesia/patología , Anestésicos Intravenosos/toxicidad , Animales , Región CA1 Hipocampal/patología , Modelos Animales de Enfermedad , Técnicas In Vitro , Aprendizaje por Laberinto/efectos de los fármacos , Técnicas de Placa-Clamp , Propofol/toxicidad , Ratas , Ratas Sprague-Dawley
5.
Neurosci Lett ; 465(1): 55-60, 2009 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-19733210

RESUMEN

AIMS: To test the hypothesis that uridine 5'-triphosphate (UTP) had a protective effect on cerebral ischemia reperfusion (IR) injury in rats. METHODS: Ischemia was induced by intraluminal suture of middle cerebral artery occlusion (MCAO). UTP solution was delivered through an indwelling tail venous catheter via microinfusion pump 30 min after the occlusion of MCA at a rate of 0.5 ml/100 g/min. Neurological deficit score (NDS) and brain water content were determined 24 h after reperfusion. Infarct volume was determined by 2,3,5-triphenyl-tetrazolium chloride (TTC) staining and magnetic resonance imaging (MRI), and nerve cell death was studied under an electron microscope. RESULTS: There was a dose-dependent relationship among 10, 30 and 90 microg/kg UTP. The 90 microg/kg UTP had the best protective effect among the 3 groups. We compared 90 microg/kg UTP group with normal saline group and found that UTP had a protective effect on cerebral IR by the results of TTC staining (15.9% vs 30.5%, P<0.01). MRI at 6, 30 and 54 h after reperfusion showed smaller infarct volume in 90 microg/kg group compared with 0 microg/kg group (283.5, 352.1, 367.45 mm(3) vs 401.36, 576.75 and 677.11 mm(3), respectively), and electron microscope showed less nerve cell death in 90 microg/kg group compared with 0 microg/kg group. CONCLUSION: UTP has a dose-dependent protective effect on cerebral IR.


Asunto(s)
Encéfalo/efectos de los fármacos , Trastornos Cerebrovasculares/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Daño por Reperfusión/tratamiento farmacológico , Uridina Trifosfato/farmacología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Muerte Celular/efectos de los fármacos , Trastornos Cerebrovasculares/metabolismo , Trastornos Cerebrovasculares/patología , Relación Dosis-Respuesta a Droga , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Imagen por Resonancia Magnética , Masculino , Microscopía Electrónica , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/administración & dosificación , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Índice de Severidad de la Enfermedad , Sales de Tetrazolio , Uridina Trifosfato/administración & dosificación , Agua/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...