Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Geriatr Nurs ; 44: 24-29, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35007867

RESUMEN

Surveys based on western populations have identified many risk factors for dysphagia in older people, but the potential risk factors consistent with the demographic characteristics of older, hospitalized Chinese patients require further study. This single-center prospective study aimed to determine the incidence of dysphagia in western China, and to develop and validate a model to predict the risk of dysphagia among older patients. A total of 343 inpatients (aged ≥ 65 years without dysphagia and cognitive impairment) were included. A score ≥ 2 on the Eating Assessment Tool-10 was defined as dysphagia. After a six-month follow-up, 70 (20.4%) patients were found to have dysphagia. The final model included age, wearing dentures, activities of daily living, cerebral vascular disease, coronary heart disease, and malignancy. The developed model has high predictive accuracy and can be easily implemented in daily practice.


Asunto(s)
Disfunción Cognitiva , Trastornos de Deglución , Actividades Cotidianas , Anciano , China/epidemiología , Disfunción Cognitiva/complicaciones , Trastornos de Deglución/epidemiología , Humanos , Estudios Prospectivos
2.
Front Physiol ; 11: 596326, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33391015

RESUMEN

Central nervous system (CNS) oxygen toxicity (CNS-OT) is a toxic reaction that appears after the inhalation of gas at an excessive oxygen partial pressure during underwater operation or hyperbaric oxygen (HBO) treatment. The mechanism of CNS-OT has not been clearly characterized. Though it has been attributed to the excessive oxidative stress induced by HBO, evidences against this hypothesis have been reported. Here we find that Forkhead box protein O3 (FoxO3a) is important for CNS-OT protection. FoxO3a knock-out (KO) mice had a shorter latency to develop convulsions and greater number of seizures within a certain period of time. The acute lung injury (ALI) induced by CNS-OT was also more severe in FoxO3a KO mice. Further analysis reveals a significant decrease in the activity of catalase (CAT), an antioxidant enzyme and a significant increase in the content of malondialdehyde (MDA), an oxidative product, in brain tissues of FoxO3a KO mice. Short-time HBO exposure could increase FoxO3a expression level and trigger its nuclear translocation. The level of nuclear localized FoxO3a peaked at 8 h after exposure. Our results demonstrate that the activity of FoxO3a is highly sensitive to HBO exposure and FoxO3a plays important roles in protecting CNS-OT. Further mechanic analysis reveals that FoxO3a protects CNS-OT via activating antioxidative signaling pathway.

3.
J Physiol Sci ; 69(5): 779-790, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31286450

RESUMEN

Intermittent hyperbaric oxygen exposure (IE-HBO) can protect the body against oxygen toxicity, but the underlying mechanisms are not very clear. Peroxiredoxin 6 (Prdx6) is a special endogenous antioxidative protein. We explored if the protective effects of IE-HBO are related to Prdx6. Mice were exposed to 280 kPa O2 for 60 min, followed by 30-min exposure to 20% O2/N2 mixture with equal pressure, repeated for six cycles. The Prdx6 protein level and non-selenium glutathione peroxidase (NSGPx) activity in the brain and lungs were then measured and the injury degree of lung and the oxidation level of brain and lung were evaluated. On this basis, the relationship between Prdx6 and IE-HBO's protection was explored. Generally, both IE-HBO and continuous exposure to HBO (CE-HBO) could increase the protein and mRNA levels of Prdx6, and such increases were more significant 24 h after cessation of exposure; moreover, the Prdx6 level of IE-HBO was higher than that of CE-HBO in both brain and lung, also more significantly 24 h after cessation of exposure. In addition, IE-HBO exposure could more effectively potentiate the activity of NSGPx and increase GSH content in brain and lung tissues. At the same time, it could reduce oxidation products in these tissues. IE-HBO could also provide protection for the lungs against injuries resulting from prolonged HBO exposure. These data showed that IE-HBO can potentiate the production and the activity of Prdx6 and consequently mitigate oxidative damages in brain and lungs. The influences of IE-HBO on Prdx6 may form an important basis for its protection against oxygen toxicity.


Asunto(s)
Oxígeno/efectos adversos , Oxígeno/metabolismo , Peroxiredoxina VI/metabolismo , Animales , Antioxidantes/metabolismo , Encéfalo/metabolismo , Glutatión Peroxidasa/metabolismo , Oxigenoterapia Hiperbárica/métodos , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo/fisiología , ARN Mensajero/metabolismo
4.
Neurotox Res ; 36(1): 193-203, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30927242

RESUMEN

The metabolism of adenosine (ADO) and nitric oxide (NO) in brain tissues is closely associated with the change of oxygen content. They have contrary effects in the onset of hyperbaric oxygen (HBO)-induced central nervous system oxygen toxicity (CNS OT): ADO can suppress the onset, while NO promotes it. We adopted the ADO-augmenting measure and NO-inhibiting measure in this study and found the combined use had a far superior preventive and therapeutic effect in protecting against CNS OT compared with the use of either measure alone. So we hypothesized that there is an interaction between ADO and NO which has an important impact on the onset of CNS OT. On this basis, we administered ADO-augmenting or ADO-inhibiting drugs to rats. After exposure to HBO, the onset of CNS OT was evaluated, followed by the measurement of NO content in brain tissues. In another experiment, rats were administered NO-augmenting or NO-inhibiting drugs. After exposure to HBO, the onset of CNS OT was evaluated, followed by measurement of the activities of ADO metabolism-related enzymes in brain tissues. The results showed that, following ADO augmentation, the content of NO and its metabolite was significantly reduced, and the onset of CNS OT significantly improved. After ADO inhibition, just the opposite was observed. NO promotion resulted in a decrease in the activity of ADO-producing enzyme, an increase in the activity of ADO-decomposing enzyme, and an aggravation in CNS OT. The above results were all reversed after an inhibition in NO content. Studies have shown that exposure to HBO has a significant impact on the content of ADO and NO in brain tissues as well as their biological effects, and ADO and NO might have an intense interaction, which might generate an important effect on the onset of CNS OT. The prophylaxis and treatment effects of CNS OT can be greatly enhanced by augmenting ADO and inhibiting NO.


Asunto(s)
Adenosina/metabolismo , Corteza Cerebral/metabolismo , Óxido Nítrico/metabolismo , Oxígeno/toxicidad , Adenosina/administración & dosificación , Adenosina Quinasa/metabolismo , Animales , Indazoles/administración & dosificación , Pulmón/patología , Masculino , Óxido Nítrico Sintasa de Tipo I/antagonistas & inhibidores , Ratas Sprague-Dawley
5.
Diving Hyperb Med ; 48(4): 235-240, 2018 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-30517956

RESUMEN

BACKGROUND: Recent studies indicated that ketone ester R,S-1,3-butanediol acetoacetate diester (BD-AcAc2) may be effective in preventing central nervous system oxygen toxicity (CNS-OT) and concomitant acute lung injury, a serious medical problem to be faced when breathing hyperbaric oxygen (HBO). This study aimed to further investigate the protective effects of BD-AcAc2 against CNS-OT and concomitant acute lung injury (ALI) in mice. METHODS: Mice were treated with BD-AcAc2 in peanut oil vehicle (2.5, 5.0 or 10.0 g·kg⁻² body weight) by gavage 20 minutes before 600 kPa HBO exposure. Control mice received the vehicle only. Seizure latency was recorded. Malondialdehyde content in brain and lung tissues, total protein level in bronchoalveolar lavage fluid (BLF) and lung water content were measured 60 minutes after the hyperbaric exposure. Histopathology of lung tissue was undertaken. RESULTS: Compared with the vehicle alone, BD-AcAc2 prolonged seizure latency in a dose-dependent manner (P < 0.01). The HBO-induced increase in brain malondialdehyde, BLF protein and lung water were significantly reduced by BD-AcAc2 (P < 0.01). CONCLUSION: Oral administration of the ketone ester BD-AcAc2 significantly protected against CNS-OT and concomitant ALI. Alleviation of oxidative stress may be one underlying mechanism providing this effect.


Asunto(s)
Acetoacetatos/uso terapéutico , Lesión Pulmonar Aguda , Encéfalo/efectos de los fármacos , Butileno Glicoles/uso terapéutico , Oxigenoterapia Hiperbárica , Acetoacetatos/farmacología , Lesión Pulmonar Aguda/tratamiento farmacológico , Animales , Oxigenoterapia Hiperbárica/efectos adversos , Ratones , Oxígeno , Ratas Sprague-Dawley , Convulsiones/tratamiento farmacológico
6.
Med Gas Res ; 7(4): 236-240, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29497483

RESUMEN

The veins are a major site of bubble formation after decompression and the lung is a target organ of bubbles. Bubble-induced inflammation has been implicated in the pathogenesis of decompression sickness (DCS). Macrophages play a central role in the inflammation, and macrophage polarization is closely related to the pathogenesis of some lung diseases. This study aimed to investigate the blood macrophage polarization in mice after decompression. BALB/c mice were exposed to hyperbaric air for 60 minutes, and rapid decompression was performed to induce DCS. Slow decompression and hyperoxia (150 kPa, 60 minutes) served as control groups, and hyperbaric oxygen (HBO; 250 kPa, 60 minutes) was employed for DCS treatment. Macrophage phenotype was determined by flow cytometry, and cytokines related to macrophage polarization were measured by enzyme-linked immunosorbent assay. Our results showed rapid decompression significantly induced the shift to M1 phenotype, which was not observed in slow decompression group, HBO and hyperoxia groups. These changes were consistent with the change in blood tumor necrosis factor α level. Moreover, any treatment could significantly increase the M2 macrophages, but blood interleukin-10 remained unchanged after different treatments. In addition, the blood and lung levels of monocyte chemoattractant protein-1 and intercellular adhesion molecule-1 increased significantly after rapid decompression, but reduced markedly after HBO treatment. Taken together, rapid decompression is able to induce the shift to M1 phenotype in blood macrophages, which may then migrate into the lung involving decompression-induced lung injury.

7.
Brain Res ; 1635: 180-9, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26806404

RESUMEN

Hyperbaric oxygen (HBO) is widely used in military operations, especially underwater missions. However, prolonged and continuous inhalation of HBO can cause central nervous system oxygen toxicity (CNS-OT), which greatly limits HBO's application. The regulation of astrocytes to the metabolism of adenosine is involved in epilepsy. In our study, we aimed to observe the effects of HBO exposure on the metabolism of adenosine in the brain. Furthermore, we aimed to confirm the possible mechanism underlying adenosine's mediation of the CNS-OT. Firstly, anesthetized rats exposed to 5 atm absolute HBO for 80 min. The concentrations of extracellular adenosine, ATP, ADP, and AMP were detected. Secondly, free-moving rats were exposed to HBO at the same pressure for 20 min, and the activities of 5'-nucleotidase and ADK in brain tissues were measured. For the mechanism studies, we observed the effects of a series of different doses of drugs related to adenosine metabolism on the latency of CNS-OT. Results showed HBO exposure could increase adenosine content by inhibiting ADK activity and improving 5'-nucleotidase activity. And adenosine metabolism during HBO exposure may be a protective response against HBO-induced CNS-OT. Moreover, the improvement of adenosine concentration, activation of adenosine A1R, or suppression of ADK and adenosine A2AR, which are involved in the prevention of HBO-induced CNS-OT. This is the first study to demonstrate HBO exposure regulated adenosine metabolism in the brain. Adenosine metabolism and adenosine receptors are related to HBO-induced CNS-OT development. These results will provide new potential targets for the termination or the attenuation of CNS-OT.


Asunto(s)
Adenosina/metabolismo , Astrocitos/metabolismo , Encéfalo/metabolismo , Oxígeno/toxicidad , 5'-Nucleotidasa/metabolismo , Adenosina/administración & dosificación , Animales , Encéfalo/efectos de los fármacos , Infusiones Intraventriculares , Masculino , Ratas , Ratas Sprague-Dawley
8.
Neuroreport ; 27(2): 73-9, 2016 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-26619231

RESUMEN

Hyperbaric oxygen (HBO) has been used widely in many underwater missions and clinical work. However, exposure to extremely high oxygen pressure may cause central nervous system oxygen toxicity (CNS-OT). The regulation of astrocyte glutamate metabolism is closely related to epilepsy. This study aimed to observe the effects of HBO exposure on glutamate metabolism in astrocytes and confirm the role of glutamate metabolism in CNS-OT. Anesthetized rats were exposed to 5 atmosphere absolute HBO for 80 min and microdialysis samples of brain interstitial fluid were continuously collected. Extracellular glutamate and glutamine concentrations were also detected. Freely moving rats were exposed to HBO of the same pressure for 20 min and glutamine synthetase (GS) activity in brain tissues was measured. Finally, we observed the effects of different doses of drugs related to glutamate metabolism on the latency of CNS-OT. Results showed that HBO exposure significantly increased glutamate content, whereas glutamine content was significantly reduced. Moreover, HBO exposure significantly reduced GS activity. Glutamate transporter-1 (GLT-1) selective antagonist ceftriaxone prolonged CNS-OT latency, whereas GLT-1 selective inhibitor dihydrokainate shortened CNS-OT latency. In summary, HBO exposure improved glutamate concentration and reduced glutamine concentration by inhibition of GS activity. GLT-1 activation also participated in the prevention of HBO-induced CNS-OT. Our research will provide a potential new target to terminate or attenuate CNS-OT.


Asunto(s)
Astrocitos/metabolismo , Encéfalo/metabolismo , Ácido Glutámico/metabolismo , Oxígeno/toxicidad , Presión del Aire , Animales , Ceftriaxona/administración & dosificación , Transportador 2 de Aminoácidos Excitadores/agonistas , Transportador 2 de Aminoácidos Excitadores/antagonistas & inhibidores , Glutamato-Amoníaco Ligasa/metabolismo , Ácido Kaínico/administración & dosificación , Ácido Kaínico/análogos & derivados , Masculino , Ratas , Ratas Sprague-Dawley
9.
CNS Neurosci Ther ; 21(12): 943-52, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26511680

RESUMEN

AIMS: This study was designed to examine the neuroprotective effects of bakkenolide-IIIa, a major novel compound extracted from the rhizome of P. trichinous. METHODS: Transient focal cerebral damage model in rats and oxygen-glucose deprivation (OGD) in cultured hippocampal neurons were performed. The amount of apoptotic neurons was determined using TUNEL assay. The expressions of Bcl-2, Bax, Akt, ERK1/2, IKKß, IκBα were measured using Western blot. The nuclear translocation and activation of NF-κB was measured using a fluorescence microscope and electrophoretic mobility shift assay (EMSA). RESULTS: Bakkenolide-IIIa (4, 8, 16 mg/kg; i.g.) was administered immediately after reperfusion could reduce the brain infarct volume, and the neurological deficit, as well as a high dose of bakkenolide-IIIa, increases the 72 h survival rate in cerebrally damaged rats. In vitro data demonstrated that bakkenolide-IIIa could increase cell viability and decrease the amount of apoptotic cells in cultured primary hippocampal neurons exposed to OGD. Bakkenolide-IIIa also dose-dependently increased the ratio of Bcl-2 to Bax. These results indicated that inhibition of apoptosis partly mediated the neuroprotection of bakkenolide-IIIa. Furthermore, bakkenolide-IIIa inhibited the phosphorylation of Akt, ERK1/2, IKKß, IκBα, and p65 in cultured hippocampal neurons exposed to OGD. Bakkenolide-IIIa not only inhibited the nuclear translocation of NF-κB in cultured neurons exposed to OGD, but also inhibited the activation of NF-κB in peri-infarct area in cerebrally damaged rats. CONCLUSION: Collectively, our findings indicated that bakkenolide-IIIa protects against cerebral damage by inhibiting AKT and ERK1/2 activation and inactivated NF-κB signaling.


Asunto(s)
Hipoxia de la Célula/efectos de los fármacos , Glucosa/deficiencia , Hipocampo/efectos de los fármacos , Ataque Isquémico Transitorio/tratamiento farmacológico , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Sesquiterpenos/farmacología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/fisiología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Hipoxia de la Célula/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Hipocampo/metabolismo , Hipocampo/patología , Infarto de la Arteria Cerebral Media , Ataque Isquémico Transitorio/metabolismo , Ataque Isquémico Transitorio/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , FN-kappa B/metabolismo , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/química , Proteínas Proto-Oncogénicas c-akt/metabolismo , Distribución Aleatoria , Ratas Sprague-Dawley , Sesquiterpenos/química
10.
Undersea Hyperb Med ; 42(2): 115-23, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26094286

RESUMEN

Decompression sickness (DCS) is a specific diving injury which sometimes may be life-threatening. Previous studies suggested that simvastatin (SIM) can protect against pathological inflammation and tissue damage. This study aimed to investigate whether SIM pretreatment could exert its beneficial effects on DCS. SIM was administered orally to adult male Sprague-Dawley rats for two weeks (2 mg/kg/day), then rats were subjected to a simulated dive at 700 kPa air pressure for 100 minutes before rapid decompression. After 30 minutes of symptom observation, lung tissue and blood samples were collected for further analysis. Compared to the vehicle-control, SIM pretreatment significantly decreased the incidence of DCS and ameliorated all parameters of pulmonary injuries, including lung dry/wet weight ratio, bronchoalveolar lavage fluid protein concentration, lung tissue malondialdehyde level and morphology. Moreover, SIM pretreatment abolished increases in systemic and pulmonary inflammation by reducing tumor necrosis factor-α levels in blood plasma and lung tissue. The results indicate that SIM may offer a novel pharmacological protection against injuries in DCS rats by inhibiting inflammatory responses. Further study is needed to understand the exact mechanisms.


Asunto(s)
Enfermedad de Descompresión/prevención & control , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Simvastatina/uso terapéutico , Adiposidad , Administración Oral , Animales , Líquido del Lavado Bronquioalveolar/química , Descompresión/métodos , Enfermedad de Descompresión/epidemiología , Enfermedad de Descompresión/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Incidencia , Inflamación/prevención & control , Lípidos/sangre , Pulmón/química , Pulmón/patología , Lesión Pulmonar/metabolismo , Lesión Pulmonar/prevención & control , Masculino , Malondialdehído/análisis , Tamaño de los Órganos , Neumonía/prevención & control , Edema Pulmonar/diagnóstico , Ratas , Ratas Sprague-Dawley , Simvastatina/administración & dosificación , Factor de Necrosis Tumoral alfa/análisis
11.
Undersea Hyperb Med ; 42(1): 49-56, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26094304

RESUMEN

This study aimed to investigate the protective effects of pravastatin on hyperbaric hyperoxia-induced lung injury (HILI). C57BL/6 mice were randomly assigned into three groups: control group, HILI group and pravastatin (Pra) group. Mice in the HILI and Pra groups were subjected to exposure to pure oxygen at 2.5 atm abs for six hours. Mice in the Pra group were intraperitoneally treated with pravastatin at 15 mg/kg. immediately after exposure. At 24 hours, the lungs were collected for HE staining, TUNEL staining and detection of lung edema, myeloperoxidase (MPO) activity and cytokines, and bronchoalveolar lavage fluid (BALF) was harvested for cell-counting. Pravastatin treatment significantly improved the pathology of the lung after HILI (reduction in thickness of alveolar septum, attenuation of lung edema, fracturing of alveolar septa and decrease in infiltrated leukocytes); reduced the number of apoptotic cells; inhibited lung MPO activity; and regulated the balance between pro-inflammatory and anti-inflammatory cytokines. Our findings suggest that pravastatin may exert a protective effect on lung injury after hyperbaric hyperoxia exposure by inhibiting inflammation.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Oxigenoterapia Hiperbárica/efectos adversos , Hiperoxia/complicaciones , Pravastatina/farmacología , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/patología , Animales , Líquido del Lavado Bronquioalveolar/citología , Quimiocina CXCL2/análisis , Etiquetado Corte-Fin in Situ , Interleucina-10/análisis , Interleucina-1beta/análisis , Pulmón/química , Masculino , Ratones , Ratones Endogámicos C57BL , Peroxidasa/análisis , Edema Pulmonar/tratamiento farmacológico
12.
Int Immunopharmacol ; 24(2): 182-190, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25523460

RESUMEN

Rubi Fructus, a traditional Chinese medicine, was considered as an anti-inflammatory agent in folk medicine. In the present study, we investigated the signalling pathways involved in the anti-inflammatory effects of goshonoside-F5 (GF5), isolated from Rubi Fructus, in peritoneal macrophages and examined its therapeutic effect in a mouse endotoxic shock model. GF5 decreased NO and PGE2 production in LPS-stimulated macrophages (IC50=3.84 and 3.16µM). This effect involved the suppression of NOS-2 and COX-2 gene expression at the transcriptional level. Examination of the effects of GF5 on NF-κB signalling demonstrated that it inhibits the phosphorylation of IκB-α and IκB-ß, blocking their degradation and the nuclear translocation of the NF-κB p65 subunit. Moreover, inhibition of MAPK signalling was also observed, and phosphorylation of p38 and JNK was suppressed in the presence of GF5. Inflammatory cytokines, including IL-6 and TNF-α, were down-regulated by this compound after activation with LPS (IC50=17.04 and 4.09µM). Additionally, GF5 (30 and 90mg/kg, i.p.) significantly reduced the circulating cytokine levels (IL-6 and TNF-α) and increased survival in a mouse model of endotoxemia. These results show that GF5 significantly inhibits the pro-inflammatory response induced by LPS, both in vitro and in vivo. Our results provide a strong pharmacological basis for further understanding the potential therapeutic role of GF5 in inflammatory disease and shed new light on the bioactivity of ent-labdane diterpene glucoside.


Asunto(s)
Antiinflamatorios/administración & dosificación , Flavonoides/administración & dosificación , Macrófagos Peritoneales/efectos de los fármacos , Medicina Tradicional China/métodos , Choque Séptico/tratamiento farmacológico , Animales , Células Cultivadas , Terapia de Inmunosupresión , Mediadores de Inflamación/metabolismo , Interleucina-6/metabolismo , Macrófagos Peritoneales/inmunología , Ratones , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Fosforilación/efectos de los fármacos , Rubus/inmunología , Choque Séptico/inmunología , Choque Séptico/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
14.
Undersea Hyperb Med ; 41(3): 183-202, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24984313

RESUMEN

Oxygen therapy is one of the most widely used clinical interventions to counteract insufficient pulmonary oxygen delivery in patients with severe lung injury. However, prolonged exposure to hyperoxia at elevated partial pressure leads to inflammation and acute lung injury. The population at risk for this condition has markedly increased with the advent of efficient systems for delivery of high concentrations of oxygen in hospitals. Thus, the therapy of hyperoxia-induced lung injury has been a focus in studies of pediatrics and pulmonary medicine. In this paper, we briefly summarized the advances in the therapies of hyperoxia-induced lung injury on the basis of its pathogenesis. We hope our summary will help provide evidence for further investigation of therapeutic measures for hyperoxia-induced lung injury.


Asunto(s)
Lesión Pulmonar Aguda/terapia , Oxigenoterapia Hiperbárica/efectos adversos , Modelos Animales , Terapia por Inhalación de Oxígeno/efectos adversos , Lesión Pulmonar Aguda/etiología , Animales , Estrés Oxidativo , Terapia por Inhalación de Oxígeno/métodos , Presión Parcial
15.
J Neurotrauma ; 31(15): 1343-53, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24716787

RESUMEN

In this study, we investigated whether nuclear factor erythroid 2-related factor 2 (Nrf2) activation in astrocytes contributes to the neuroprotection induced by a single hyperbaric oxygen preconditioning (HBO-PC) against spinal cord ischemia/reperfusion (SCIR) injury. In vivo: At 24 h after a single HBO-PC at 2.5 atmospheres absolute for 90 min, the male ICR mice underwent SCIR injury by aortic cross-clamping surgery and observed for 48 h. HBO-PC significantly improved hindlimb motor function, reduced secondary spinal cord edema, ameliorated the reactivity of spinal motor-evoked potentials, and slowed down the process of apoptosis to exert neuroprotective effects against SCIR injury. At 12 h or 24 h after HBO-PC without aortic cross-clamping surgery, Western blot, enzyme-linked immunosorbent assay, realtime-polymerase chain reaction and double-immunofluorescence staining were used to detect the Nrf2 activity of spinal cord tissue, such as mRNA level, protein content, DNA binding activity, and the expression of downstream gene, such as glutamate-cysteine ligase, γ-glutamyltransferase, multidrug resistance protein 1, which are key proteins for intracellular glutathione synthesis and transit. The Nrf2 activity and downstream genes expression were all enhanced in normal spinal cord with HBO-PC. Glutathione content of spinal cord tissue with HBO-PC significantly increased at all time points after SCIR injury. Moreover, Nrf2 overexpression mainly occurs in astrocytes. In vitro: At 24 h after HBO-PC, the primary spinal astrocyte-neuron co-cultures from ICR mouse pups were subjected to oxygen-glucose deprivation (OGD) for 90 min to simulate the ischemia-reperfusion injury. HBO-PC significantly increased the survival rate of neurons and the glutathione content in culture medium, which was mainly released from asctrocytes. Moreover, the Nrf2 activity and downstream genes expression induced by HBO-PC were mainly enhanced in astrocytes, but not in neurons. In conclusion, our findings demonstrated that spinal cord ischemic tolerance induced by HBO-PC may be mainly related to Nrf2 activation in astrocytes.


Asunto(s)
Astrocitos/metabolismo , Oxigenoterapia Hiperbárica/métodos , Precondicionamiento Isquémico/métodos , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Western Blotting , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Ratones Endogámicos ICR , Reacción en Cadena en Tiempo Real de la Polimerasa , Recuperación de la Función , Traumatismos de la Médula Espinal/metabolismo , Isquemia de la Médula Espinal/metabolismo
16.
PLoS One ; 9(1): e85967, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24465817

RESUMEN

OBJECTIVE: Hyperbaric oxygen (HBO) preconditioning (HBO-PC) has been testified to have protective effects on spinal cord injury (SCI). However, the mechanisms remain enigmatic. The present study aimed to explore the effects of HBO-PC on primary rat spinal neurons against oxidative injury and oxygen-glucose deprivation (OGD) and the relationship with heat shock proteins (HSPs). METHODS: Primary rat spinal neurons after 7 days of culture were used in this study. HSPs were detected in rat spinal neurons following a single exposure to HBO at different time points by Western blot. Using lactate dehydrogenase release assay and cell counting kit-8 assay, the injuries induced by hydrogen peroxide (H2O2) insult or OGD were determined and compared among neurons treated with HBO-PC with or without HSP inhibitors. RESULTS: The results of Western blot showed that HSP27, HSP70 and HSP90 have a slight but not significant increase in primary neurons following HBO exposure. However, HSP32 expression significantly increased and reached highest at 12 h following HBO exposure. HBO-PC significantly increased the cell viability and decreased the medium lactate dehydrogenase content in cultures treated with H2O2 or OGD. Pretreatment with zinc protoporphyrin IX, a specific inhibitor of HSP32, significantly blocked the protective effects of HBO-PC. CONCLUSIONS: These results suggest that HBO-PC could protect rat spinal neurons in vitro against oxidative injury and OGD mostly by up-regulating of HSP32 expression.


Asunto(s)
Glucosa/deficiencia , Hemo Oxigenasa (Desciclizante)/metabolismo , Oxigenoterapia Hiperbárica , Neuronas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Oxígeno/farmacología , Columna Vertebral/patología , Regulación hacia Arriba/efectos de los fármacos , Animales , Células Cultivadas , Hipoxia/patología , Neuronas/efectos de los fármacos , Ratas , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología , Columna Vertebral/efectos de los fármacos , Factores de Tiempo
17.
CNS Neurol Disord Drug Targets ; 13(5): 874-84, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24040794

RESUMEN

Total bakkenolides is the major component of the rhizome of Petasites trichinous Franch.. In this study, we investigated its neuroprotective effects in a rat transient focal cerebral ischemia-reperfusion model, and in an in vitro cerebral ischemia model, oxygen-glucose deprivation of cultured nerve cells. Oral administration of total bakkenolides immediately after reperfusion at doses of 5, 10 and 20 mg/kg markedly reduced brain infarct volume and neurological deficits. Total bakkenolides significantly attenuated cell death and apoptosis in primarily cultured neurons subject to 1-h hypoxia followed by 24-h reoxygenation. Morphologic observations directly confirmed its protective effect on neurons. We also demonstrated that total bakkenolides could inhibit nuclear factor-κB (NF-κB) activation by blocking the classic activation pathway through suppression of phosphorylation of IκB-kinase complex, NF-κB/p65 and inhibitor protein IκB, inducing nuclear translocation of NF-κB/p65 and degradation of IκB. Further, total bakkenolides inhibited the activation of Akt and the extracellular signal-regulated kinase 1/2, two important upstream activators of NF-κB. In conclusion, our results provide a strong pharmacological basis for further understanding the potential therapeutic role of total bakkenolides in cerebral ischemic disease and shed new light on its neuroprotective mechanism.


Asunto(s)
Ataque Isquémico Transitorio/prevención & control , FN-kappa B/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Sesquiterpenos/uso terapéutico , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Infarto Encefálico/etiología , Infarto Encefálico/prevención & control , Células Cultivadas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Hipocampo/citología , Etiquetado Corte-Fin in Situ , Ataque Isquémico Transitorio/complicaciones , Masculino , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Enfermedades del Sistema Nervioso/etiología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Sprague-Dawley , Sesquiterpenos/química , Sesquiterpenos/farmacología
18.
Undersea Hyperb Med ; 41(6): 573-8, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25562948

RESUMEN

Hyperbaric oxygen therapy is one of the most widely used clinical interventions to counteract insufficient pulmonary oxygen delivery in patients with severe lung injury. However, prolonged exposure to hyperoxia leads to inflammation and acute lung injury. This study aimed to investigate the protective effect of hydrogen sulfide on hyperbaric hyperoxia-induced lung injury. Rats were intraperitoneally treated with sodium hydrosulphide (NaHS) at 28 µmol/kg immediately before hyperoxia exposure and then exposed to pure oxygen at 2.5 atmospheres absolute (atm abs) with continuous ventilation for six hours, Immediately after hyperoxia exposure, rats were sacrificed via anesthesia. The bronchoalveolar lavage fluid (BALF) was harvested for the detection of protein concentration and IL-1 content, and the lungs were collected for HE staining, TUNEL staining and detection of wet/dry weight ratio. Our results showed hyperbaric hyperoixa exposure could significantly damage the lung (HE staining), increase the protein and IL-13 in the BALF, elevate the wet/dry Weight ratio and raise the TUNEL positive cells. However, pre-treatment with hydrogen sulfide improved the lung morphology, reduced the TUNEL positive cells and attenuated the lung inflammation (reduction in IL-13 of BALF and HE staining). Taken together, our findings indicate that hydrogen sulfide pretreatment may exert protective effects on hyperbaric hyperoxia-induced lung injury.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Gasotransmisores/uso terapéutico , Sulfuro de Hidrógeno/uso terapéutico , Oxigenoterapia Hiperbárica/efectos adversos , Lesión Pulmonar Aguda/etiología , Animales , Antracenos , Líquido del Lavado Bronquioalveolar/química , Etiquetado Corte-Fin in Situ , Inyecciones Intraperitoneales , Interleucina-1beta/análisis , Pulmón/efectos de los fármacos , Pulmón/patología , Masculino , Proteínas/análisis , Ratas Sprague-Dawley , Sulfuros/farmacología
19.
Exp Biol Med (Maywood) ; 238(1): 12-22, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23479759

RESUMEN

Decompression sickness (DCS) is a major concern in diving and space walk. Hyperbaric oxygen (HBO) preconditioning has been proved to enhance tolerance to DCS via nitric oxide. Heat-shock protein (HSP) 70 was also found to have protective effects against DCS. We hypothesized that the beneficial effects of HBO preconditioning on DCS was related to levels of elevated HSP70. HSPs (70, 27 and 90) expressed in tissues of spinal cord and lung in rats was detected at different time points following HBO exposure by Western blot. HSP27 and HSP90 showed a slight but not significant increase after HBO. HSP70 increased and reached highest at 18 h following exposure before decreasing. Then rats were exposed to HBO and subjected to simulated air dive and rapid decompression to induce DCS 18 h after HBO. The severity of DCS, along with levels of HSP70 expression, as well as the extent of oxidative and apoptotic parameters in the lung and spinal cord were compared among different groups of rats pretreated with HBO, HBO plus NG-nitro-l-arginine-methyl ester (l-NAME), HBO plus quercetin or normobaric air. HBO preconditioning significantly reduced the morbidity of DCS (from 66.7% to 36.7%), reduced levels of oxidation (malondialdehyde, 8-hydroxyguanine and hydrogen peroxide) and apoptosis (caspase-3 and -9 activities and the number of apoptotic cells). l-NAME or quercetin eliminated most of the beneficial effects of HBO on DCS, and counteracted the stimulation of HSP70 by HBO. Bubbles in pulmonary artery were detected using ultrasound imaging to observe the possible effect of HBO preconditioning on DCS bubble formation. The amounts of bubbles in rats pretreated with HBO or air showed no difference. These results suggest that HSP70 was involved in the beneficial effects of HBO on DCS in rats, suspected be by the antioxidation and antiapoptosis effects.


Asunto(s)
Enfermedad de Descompresión/patología , Enfermedad de Descompresión/fisiopatología , Proteínas HSP70 de Choque Térmico/metabolismo , Oxigenoterapia Hiperbárica , Animales , Western Blotting , Perfilación de la Expresión Génica , Pulmón/química , Pulmón/patología , Masculino , Ratas , Ratas Sprague-Dawley , Índice de Severidad de la Enfermedad , Médula Espinal/química , Médula Espinal/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...