Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Med Gas Res ; 10(1): 30-36, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32189667

RESUMEN

Carbon monoxide (CO) has been the leading cause of poisoning mortality in many countries and hyperbaric oxygen (HBO) is a widely accepted treatment for CO poisoning. However, some patients with CO poisoning will still develop neurocognitive sequelae regardless of HBO therapy, which can persist since CO poisoning or be present days to weeks after a recovery from CO poisoning. HBO has been used in the prevention and treatment of neurocognitive sequelae after CO poisoning, and some mechanisms are also proposed for the potential neuroprotective effects of HBO on the neurocognitive impairment after CO poisoning, but there is still controversy on the effectiveness of HBO on neurocognitive sequelae after CO poisoning. In this paper, we briefly introduce the neurocognitive sequelae after CO poisoning, summarize the potential predictive factors of neurocognitive sequelae, and discuss the use of HBO in the treatment and prevention of neurocognitive sequelae after CO poisoning.


Asunto(s)
Intoxicación por Monóxido de Carbono/complicaciones , Intoxicación por Monóxido de Carbono/terapia , Oxigenoterapia Hiperbárica/efectos adversos , Trastornos Neurocognitivos/complicaciones , Humanos
2.
J Neuroimmunol ; 294: 6-13, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27138092

RESUMEN

Multiple sclerosis (MS) is a chronic autoimmune demyelinating disease of the central nervous system (CNS). The high costs, inconvenient administration, and side effects of current Food and Drug Administration (FDA)-approved drugs often lead to poor adherence to the long-term treatment of MS. Molecular hydrogen (H2) has been reported to exhibit anti-oxidant, anti-apoptotic, anti-inflammatory, anti-allergy, and anti-cancer effects. In the present study, we explored the prophylactic and therapeutic effects of hydrogen-rich water (HRW) on the progress of experimental autoimmune encephalomyelitis (EAE), the animal model for MS. We found that prophylactic administration of both 0.36mM and 0.89mM HRW was able to delay EAE onset and reduce maximum clinical scores. Moreover, 0.89mM HRW also reduced disease severity, CNS infiltration, and demyelination when administered after the onset of disease. Furthermore, HRW treatment prevented infiltration of CD4(+) T lymphocytes into the CNS and inhibited Th17 cell development without affecting Th1 cell populations. Because HRW is non-toxic, inexpensive, easily administered, and can readily cross the blood-brain barrier, our experiments suggest that HRW may have great potential in the treatment of MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/complicaciones , Hidrógeno/uso terapéutico , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Enfermedades del Sistema Nervioso/etiología , Recuperación de la Función/efectos de los fármacos , Análisis de Varianza , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Citometría de Flujo , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Ratones , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito/inmunología , Glicoproteína Mielina-Oligodendrócito/toxicidad , Enfermedades del Sistema Nervioso/inmunología , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/toxicidad , Estadísticas no Paramétricas , Agua
3.
Sci Rep ; 6: 23742, 2016 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-27026206

RESUMEN

Emerging evidence suggests that neuroinflammation and oxidative stress may be major contributors to major depressive disorder (MDD). Patients or animal models of depression show significant increase of proinflammatory cytokine interleukin-1ß (IL-1ß) and oxidative stress biomarkers in the periphery or central nervous system (CNS). Recent studies show that hydrogen selectively reduces cytotoxic oxygen radicals, and hydrogen-rich saline potentially suppresses the production of several proinflammatory mediators. Since current depression medications are accompanied by a wide spectrum of side effects, novel preventative or therapeutic measures with fewer side effects might have a promising future. We investigated the effects of drinking hydrogen-rich water on the depressive-like behavior in mice and its underlying mechanisms. Our study show that hydrogen-rich water treatment prevents chronic unpredictable mild stress (CUMS) induced depressive-like behavior. CUMS induced elevation in IL-1ß protein levels in the hippocampus, and the cortex was significantly attenuated after 4 weeks of feeding the mice hydrogen-rich water. Over-expression of caspase-1 (the IL-1ß converting enzyme) and excessive reactive oxygen species (ROS) production in the hippocampus and prefrontal cortex (PFC) was successfully suppressed by hydrogen-rich water treatment. Our data suggest that the beneficial effects of hydrogen-rich water on depressive-like behavior may be mediated by suppression of the inflammasome activation resulting in attenuated protein IL-1ß and ROS production.


Asunto(s)
Trastorno Depresivo Mayor/tratamiento farmacológico , Hidrógeno/administración & dosificación , Administración Oral , Animales , Caspasa 1/metabolismo , Trastorno Depresivo Mayor/metabolismo , Evaluación Preclínica de Medicamentos , Hipocampo/efectos de los fármacos , Hipocampo/enzimología , Masculino , Ratones Endogámicos BALB C , Estrés Oxidativo , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/enzimología , Especies Reactivas de Oxígeno/metabolismo , Estrés Psicológico/tratamiento farmacológico , Agua/administración & dosificación
4.
Chin Med J (Engl) ; 126(20): 3904-9, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24157154

RESUMEN

BACKGROUND: Hyperbaric oxygen preconditioning (HBO) is a new method of ischemia preconditioning. In this study, we examined its effects on skin flap survival and the mechanisms involved. METHODS: Thirty-six rats were divided into three groups: HBO preconditioning, control, and sham groups. An extended epigastric adipocutaneous flap based on the right superficial epigastric artery and vein was raised. A 3-hour period of flap ischemia was induced by clamping the pedicle vessels with a microvascular clamp. At the end of ischemia induction, the clamp was removed and the flap was resutured. Rats in the HBO preconditioning group were treated with HBO four times before surgery. Microcirculation in the skin flap was measured on postoperative days 1, 3 and 5. The size of the flap was measured on postoperative day 5, before the animals were sacrificed. Samples of the skin flap were prepared and stained with hematoxylin and eosin. The levels of tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, and IL-6 in the flap samples were measured. RESULTS: Surviving flap size was significantly higher in the HBO preconditioning group compared with controls, with a reduced inflammatory response and increased perfusion. IL-1, TNF-α, and IL-6 levels in the HBO preconditioning group were lower than in controls. CONCLUSIONS: HBO preconditioning improved flap survival in this ischemia-reperfusion rat model. The mechanisms responsible for this effect may relate to attenuation of the inflammatory response and increased flap perfusion following HBO preconditioning.


Asunto(s)
Oxigenoterapia Hiperbárica/métodos , Isquemia/cirugía , Colgajos Quirúrgicos , Animales , Supervivencia de Injerto , Masculino , Microcirculación/fisiología , Ratas , Ratas Sprague-Dawley , Piel
6.
BMC Neurosci ; 13: 47, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22587664

RESUMEN

BACKGROUND: Increasing experimental and clinical data indicate that early brain injury (EBI) after subarachnoid hemorrhage (SAH) largely contributes to unfavorable outcomes, and it has been proved that EBI following SAH is closely associated with oxidative stress and brain edema. The present study aimed to examine the effect of hydrogen, a mild and selective cytotoxic oxygen radical scavenger, on oxidative stress injury, brain edema and neurology outcome following experimental SAH in rabbits. RESULTS: The level of MDA, caspase-12/3 and brain water content increased significantly at 72 hours after experimental SAH. Correspondingly, obvious brain injury was found in the SAH group by terminal deoxynucleotidyl transferase-mediated uridine 5'-triphosphate-biotin nick end-labeling (TUNEL) and Nissl staining. Similar results were found in the SAH+saline group. In contrast, the upregulated level of MDA, caspase-12/3 and brain edema was attenuated and the brain injury was substantially alleviated in the hydrogen treated rabbits, but the improvement of neurology outcome was not obvious. CONCLUSION: The results suggest that treatment with hydrogen in experimental SAH rabbits could alleviate brain injury via decreasing the oxidative stress injury and brain edema. Hence, we conclude that hydrogen possesses the potential to be a novel therapeutic agent for EBI after SAH.


Asunto(s)
Edema Encefálico/prevención & control , Lesiones Encefálicas/prevención & control , Hidrógeno/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Análisis de Varianza , Animales , Edema Encefálico/etiología , Lesiones Encefálicas/etiología , Caspasa 12/metabolismo , Caspasa 3/metabolismo , Muerte Celular/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Modelos Animales de Enfermedad , Etiquetado Corte-Fin in Situ , Examen Neurológico , Conejos , Hemorragia Subaracnoidea/complicaciones , Agua/metabolismo
7.
Clin Exp Pharmacol Physiol ; 38(10): 675-82, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21711378

RESUMEN

1. Heme oxygenase 1 (HO-1) has been shown to play a pivotal role in the maintenance of cellular homeostasis when the liver undergoes sublethal stress, such as ischaemia-reperfusion (I/R) injury. In the present study, we investigated the protective role of HO-1 in hyperbaric oxygen (HBO) preconditioning against liver injury after I/R. 2. A total hepatic ischaemia (30 min) and reperfusion (60 min) injury model in rats was used in the present study. Preconditioned groups were exposed to HBO 24 h prior to the induction of I/R injury. Other groups were injected with zinc protoporphyrin IX (ZnPP) intraperitoneally 1 h before I/R to inhibit HO-1 activity. At the end of the reperfusion period, blood and liver samples were collected for the analysis of liver injury markers, morphological changes, and HO-1 expression and activity in the liver. 3. In untreated rats, I/R induced an increase in hepatic injury markers, such as plasma transaminases, inflammatory cytokines (tumour necrosis factor-α and interleukin-1ß), and tissue malondialdehyde. However, HBO preconditioning attenuated the I/R-induced increases in these hepatic injury markers, and prevented both the necrosis and apoptosis of hepatocytes induced by I/R injury. Furthermore, HBO preconditioning significantly increased HO-1 mRNA and protein levels in the liver. In rats in which HO-1 activity had been inhibited with ZnPP pretreatment, the protective effects of HBO preconditioning against I/R injury were abolished. 4. In conclusion, HBO preconditioning can protect the liver against I/R injury and it appears that this effect might be mediated by the induction of HO-1.


Asunto(s)
Hemo-Oxigenasa 1/metabolismo , Oxigenoterapia Hiperbárica/métodos , Precondicionamiento Isquémico/métodos , Hepatopatías/metabolismo , Hepatopatías/prevención & control , Protoporfirinas/farmacología , Daño por Reperfusión/prevención & control , Animales , Apoptosis/efectos de los fármacos , Modelos Animales de Enfermedad , Hemo-Oxigenasa 1/antagonistas & inhibidores , Humanos , Oxigenoterapia Hiperbárica/estadística & datos numéricos , Interleucina-1beta/sangre , Hepatopatías/patología , Masculino , Malondialdehído/metabolismo , Necrosis/prevención & control , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Transaminasas/sangre , Factor de Necrosis Tumoral alfa/sangre
8.
Undersea Hyperb Med ; 37(5): 271-80, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20929184

RESUMEN

Central nervous system oxygen toxicity, which occurs during diving and hyperbaric oxygen treatment, can lead to very dangerous situations, and it is of great importance to explore its mechanisms. We have speculated that cerebral blood flow plays a pivotal role in its occurrence. Except for acting as an anticonvulsant in clinical applications, acetazolamide is also a vasodilator used in both clinical and laboratory settings. In this study, when acetazolamide from 5 to 500 ug/kg body weight was administered by intracerebroventricular injection, the latency of central nervous system oxygen toxicity detected by electroencephalogram recording in rats subjected to hyperbaric oxygen at 6 atmospheres absolute was prolonged significantly. On the contrary, when the dose of intracerebroventricular injection achieved 5,000 ug/kg body weight, acetazolamide shortened the latency significantly. Intraperitoneal injection of acetazolamide more than 7.5 mg/kg body weight also shortened the latency significantly. Results also showed both intracerebroventricular injection of acetazolamide at a dose of 5,000 ug/kg body weight and intraperitoneal injection at dose of 7.5 mg/kg body weight inhibited the activity of carbonic anhydrase and increased the cerebral blood flow significantly, which helped aggravate oxidation damage and resulted in increased MDA and impaired glutathione peroxidase in brain tissue. But intracerebroventricular injection of acetazolamide at 5 ug/kg body weight had no effect on MDA and glutathione peroxidase, though it inhibited the activity of carbonic anhydrase. These observations indicated acetazolamide covers bidirectional influences on central nervous system oxygen toxicity. Within local brain tissue, especially neurons, it could exert its anticonvulsive effect on the central nervous system at low doses. On the other hand, under high doses, it would display its convulsive-hastening effect through increasing cerebral blood flow to aggravate the oxidation state of brain tissues and exacerbate central nervous system oxygen toxicity when subjected to hyperbaric oxygen. Blood flow of brain plays a pivotal role in central nervous system oxygen toxicity.


Asunto(s)
Acetazolamida/farmacología , Encéfalo/efectos de los fármacos , Inhibidores de Anhidrasa Carbónica/farmacología , Anhidrasas Carbónicas/metabolismo , Circulación Cerebrovascular/efectos de los fármacos , Oxígeno/toxicidad , Acetazolamida/administración & dosificación , Aldehídos/metabolismo , Animales , Anticonvulsivantes/administración & dosificación , Anticonvulsivantes/farmacología , Cámaras de Exposición Atmosférica , Presión Atmosférica , Peso Corporal , Encéfalo/irrigación sanguínea , Encéfalo/fisiología , Inhibidores de Anhidrasa Carbónica/administración & dosificación , Anhidrasas Carbónicas/análisis , Sistema Nervioso Central/efectos de los fármacos , Circulación Cerebrovascular/fisiología , Relación Dosis-Respuesta a Droga , Electrodos Implantados , Electroencefalografía , Oxigenoterapia Hiperbárica , Inyecciones Intraperitoneales , Inyecciones Intraventriculares , Masculino , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Vasodilatadores/administración & dosificación , Vasodilatadores/farmacología
9.
Undersea Hyperb Med ; 37(3): 173-80, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20568547

RESUMEN

Divers are at risk of decompression sickness (DCS) when the ambient pressure decrease exceeds a critical threshold. Hyperbaric oxygen (HBO2) preconditioning has been used to prevent various injuries, but the protective effect on DCS has not been well explored. To investigate the prophylactic effect of HBO2 on DCS, rats were pretreated with HBO2 (250 kPa-60 minutes) (all the pressures described here are absolute pressure) for 18 hours before a simulated air dive (700 kPa-100 minutes) with fast decompression to the surface at the rate of 200 kPa/min (n=33). During the following 30 minutes, the rats walked in a 3 m/minute rotating cage and were monitored for signs of DCS. The control rats were pretreated with normobaric air (n=30), normoxic hyperbaric nitrox (250 kPa, 8.4% O2) (n=13), or N(G)-nitro-L-arginine methyl ester (L-NAME) 30 minutes before HBO2 exposure (n=13). Nitric oxide (NO) levels were recorded immediately and 18 hours after HBO2 exposure in the brain and spinal cord. The incidence of DCS in rats pretreated with HBO2 was 30.3%, which was significantly lower than those treated with normobaric air (63.3%) (p<0.05) or hyperbaric nitrox (61.5%) (p<0.05). The onset time of DCS of the rats pretreated with HBO2 was significantly delayed compared with those treated with air (p<0.05). L-NAME nullified the HBO2 preconditioning effect. HBO2 increased NO level in the rat brain and spinal cord right after exposure; this effect was inhibited by L-NAME. Taken together, HBO2 preconditioning reduced the incidence of DCS in rats, and NO was involved in the prophylactic effect.


Asunto(s)
Enfermedad de Descompresión/prevención & control , Oxigenoterapia Hiperbárica/métodos , Óxido Nítrico/metabolismo , Animales , Encéfalo/metabolismo , Enfermedad de Descompresión/metabolismo , Inhibidores Enzimáticos/administración & dosificación , Masculino , Actividad Motora/fisiología , NG-Nitroarginina Metil Éster/administración & dosificación , Óxido Nítrico/análisis , Nitrógeno/administración & dosificación , Oxígeno/administración & dosificación , Ratas , Ratas Sprague-Dawley , Médula Espinal/metabolismo , Factores de Tiempo
10.
Liver Int ; 30(7): 958-68, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20492513

RESUMEN

BACKGROUND: Hydrogen selectively reduces levels of hydroxyl radicals and alleviates acute oxidative stress in many models. Hydrogen-rich saline provides a high concentration of hydrogen that can be easily and safely applied. AIMS: In this study, we investigated the effects of hydrogen-rich saline on the prevention of liver injury induced by obstructive jaundice in rats. METHODS: Male Sprague-Dawley rats (n=56) were divided randomly into four experimental groups: sham operated, bile duct ligation (BDL) plus saline treatment [5 ml/kg, intraperitoneal (i.p.)], BDL plus low-dose hydrogen-rich saline treatment (5 ml/kg, i.p.) and BDL plus high-dose hydrogen-rich saline treatment (10 ml/kg, i.p.). RESULTS: The liver damage was evaluated microscopically 10 days after BDL. Serum alanine aminotransferase and aspartate aminotransferase levels, tissue malondialdehyde content, myeloperoxidase activity, tumour necrosis factor-alpha, interleukin (IL)-1beta, IL-6 and high-mobility group box 1 levels were all increased significantly by BDL. Hydrogen-rich saline reduced levels of these markers and relieved morphological liver injury. Additionally, hydrogen-rich saline markedly increased the activities of anti-oxidant enzymes superoxide dismutase and catalase and downregulated extracellular signal-regulated protein kinase (ERK)1/2 activation. CONCLUSIONS: Hydrogen-rich saline attenuates BDL-induced liver damage, possibly by the reduction of inflammation and oxidative stress and the inhibition of the ERK1/2 pathway.


Asunto(s)
Antiinflamatorios/farmacología , Antioxidantes/farmacología , Hidrógeno/farmacología , Ictericia Obstructiva/tratamiento farmacológico , Hepatopatías/prevención & control , Hígado/efectos de los fármacos , Cloruro de Sodio/farmacología , Alanina Transaminasa/sangre , Animales , Antiinflamatorios/administración & dosificación , Antioxidantes/administración & dosificación , Aspartato Aminotransferasas/sangre , Catalasa/metabolismo , Citoprotección , Modelos Animales de Enfermedad , Endotoxinas/sangre , Proteína HMGB1/metabolismo , Hidrógeno/administración & dosificación , Inyecciones Intraperitoneales , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Ictericia Obstructiva/complicaciones , Ictericia Obstructiva/metabolismo , Ictericia Obstructiva/patología , Hígado/metabolismo , Hígado/patología , Hepatopatías/etiología , Hepatopatías/metabolismo , Hepatopatías/patología , Masculino , Malondialdehído/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Peroxidasa/metabolismo , Ratas , Ratas Sprague-Dawley , Cloruro de Sodio/administración & dosificación , Superóxido Dismutasa/metabolismo , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo
11.
J Appl Physiol (1985) ; 104(4): 1185-91, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18174394

RESUMEN

We studied the effect of hyperbaric oxygen (HBO) preconditioning on the molecular mechanisms of neuroprotection in a rat focal cerebral ischemic model. Seventy-two male Sprague-Dawley rats were pretreated with HBO (100% O(2), 2 atmospheres absolute, 1 h once every other day for 5 sessions) or with room air. In experiment 1, HBO-preconditioned rats and matched room air controls were subjected to focal cerebral ischemia or sham surgery. Postinjury motor parameters and infarction volumes of HBO-preconditioned rats were compared with those of controls. In experiment 2, HBO-preconditioned rats and matched room air controls were killed at different time points. Brain levels of hypoxia-inducible factor-1alpha (HIF-1alpha) and its downstream target gene erythropoietin (EPO) analyzed by Western blotting and RT-PCR as well as HIF-1alpha DNA-binding and transcriptional activities were determined in the ipsilateral hemisphere. HBO induced a marked increase in the protein expressions of HIF-1alpha and EPO and the activity of HIF-1alpha, as well as the expression of EPO mRNA. HBO preconditioning dramatically improved the neurobehavioral outcome at all time points (3.0 +/- 2.1 vs. 5.6 +/- 1.5 at 4 h, 5.0 +/- 1.8 vs. 8.8 +/- 1.4 at 8 h, 6.4 +/- 1.8 vs. 9.7 +/- 1.3 at 24 h; P < 0.01, respectively) and reduced infarction volumes (20.7 +/- 4.5 vs. 12.5 +/- 3.6%, 2,3,5-Triphenyltetrazolium chloride staining) after cerebral ischemia. This observation indicates that the neuroprotection induced by HBO preconditioning may be mediated by an upregulation of HIF-1alpha and its target gene EPO.


Asunto(s)
Isquemia Encefálica/terapia , Eritropoyetina/biosíntesis , Oxigenoterapia Hiperbárica , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Precondicionamiento Isquémico , Animales , Conducta Animal/efectos de los fármacos , Western Blotting , Isquemia Encefálica/fisiopatología , Infarto Cerebral/patología , Infarto Cerebral/prevención & control , Circulación Cerebrovascular/fisiología , ADN/biosíntesis , ADN/genética , ADN/metabolismo , Eritropoyetina/genética , Miembro Anterior/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Masculino , Fármacos Neuroprotectores , Oxígeno/farmacología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/prevención & control , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sales de Tetrazolio , Regulación hacia Arriba/genética , Regulación hacia Arriba/fisiología
12.
Aviat Space Environ Med ; 78(4): 368-73, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17484337

RESUMEN

INTRODUCTION: The precise action of the immunological effects associated with hyperbaric exposure is poorly understood. This study's goal was to clarify the effects and etiology of deep air dives on the immune response. METHODS: Sprague-Dawley rats were exposed to 7-ATA air for 60 min twice daily for 3 consecutive days. Several markers of immune function, the degree of stress, and oxidative stress following or during the exposures were determined. Rats exposed to 1.47-ATA oxygen or 7-ATA nitrox (0.21-ATA oxygen + 6.79-ATA nitrogen) were taken as controls. RESULTS: Peripheral lymphocytes and CD3+ and CD4+CD3+ subsets in peripheral blood and spleen, plasma interleukin-2 level, and the responses of splenic lymphocytes to concanavalin A all decreased, antioxidant enzyme activities and the concentration of reduced glutathione both decreased, while the level of malondialdehyde increased following hyperbaric air exposures. All changes returned to normal in 3-5 d. Similar changes were observed following exposures to 1.47-ATA oxygen, but not to normoxic nitrox. Plasma levels of adrenocorticotropic hormone and corticosterone increased after one exposure and recovered to normal levels after three exposures in rats treated with either hyperbaric or normobaric air. Pretreatment of the animals with N-acetylcysteine, a potent free radical scavenger and antioxidant attenuated the effects of hyperbaric air on immune and antioxidant systems. CONCLUSIONS: These results are consistent with the hypothesis that repetitive exposure to 7-ATA air has a temporary immunosuppressive effect on rats, which is related to oxidative stress induced by the high partial pressure of oxygen in breathing gas.


Asunto(s)
Medicina Aeroespacial , Presión Atmosférica , Descompresión/efectos adversos , Oxigenoterapia Hiperbárica/efectos adversos , Sistema Inmunológico , Terapia de Inmunosupresión , Narcosis por Gas Inerte/etiología , Estrés Oxidativo , Animales , Masculino , Ratas , Ratas Sprague-Dawley , Linfocitos T
13.
Brain Res ; 1153: 204-13, 2007 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-17462608

RESUMEN

It has been established that hyperbaric oxygen (HBO) treatment reduces brain edema, decreases infarct volume, contributes to neurological functional recovery and suppresses apoptosis in suture-induced focal cerebral ischemic animal models. In the present study, we evaluated the therapeutic effect of HBO in an endothelin-1-induced focal cerebral ischemia in rats and explored the associated mechanisms of HBO-induced brain protection. One hundred twenty male Sprague-Dawley rats (280 to 320 g) were randomly assigned to sham, focal cerebral ischemia and focal cerebral ischemia treated with HBO groups. Brain water content, neurological function, morphology and molecular biological markers were assessed. HBO (100% O2, 2.5 atmosphere absolute for 2 h) was initiated at 1 h after focal cerebral ischemia. Rats were killed at 24 h to harvest tissues for Western blot or for histology. In HBO-treated animals, an enhanced ratio of Bcl-2 and Bax and a reduced expression of hypoxia-inducible factor-1alpha (HIF-1alpha) in the hippocampus after focal cerebral ischemia were observed. These results indicate that HBO provides brain protection that is probably associated with the inhibition of HIF-1alpha and the elevation of Bcl-2.


Asunto(s)
Isquemia Encefálica/terapia , Endotelina-1 , Oxigenoterapia Hiperbárica/métodos , Análisis de Varianza , Animales , Edema Encefálico/etiología , Edema Encefálico/terapia , Infarto Encefálico/etiología , Infarto Encefálico/terapia , Isquemia Encefálica/inducido químicamente , Isquemia Encefálica/complicaciones , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Sprague-Dawley , Proteína X Asociada a bcl-2/metabolismo
14.
Sheng Li Xue Bao ; 56(2): 158-62, 2004 Apr 25.
Artículo en Chino | MEDLINE | ID: mdl-15127124

RESUMEN

The purpose of the present study was to explore the relation between the modulation of cerebral blood flow and the latency of hyperbaric oxygen-induced convulsion. There were two parts in this study. First, the effect of acetazolamide on the latency of hyperbaric oxygen-induced convulsion was observed. 32 Sprague-Dawley (SD) rats were randomly divided into four groups: the acetazolamide 200, 20, 2 mg/kg body weight and normal saline (NS) group. The animals were given intraperitoneally acetazolamide or NS, respectively, before being exposed to the pressure of 6 ATA (absolute atmosphere) of pure oxygen. The time from exposure to the onset of seizure (clonic-tonic convulsion) was recorded for each animal according to behavioral observation. Second, the changes in maleic dialdehyde (MDA) and the activity of glutathione peroxidase (GSH-PX) were measured after acetazolamide treatment. 40 SD rats were randomly divided into five groups: NS group, 6 min with NS group, 6 min with acetazolamide group, 16 min with NS group, and 16 min with acetazolamide group. The dose of acetazolamide was 20 mg/kg body weight. After injection of NS or acetazolamide, the animals were subjected to the pressure of 6 ATA of pure oxygen in respect to its time course group. The rats were decapitated and the cortex, hippocampus, and striatum of brains were dissected and homogenized. The content of MDA and the activity of GSH-PX in these tissues were determined. We found that (1) there was a significant difference in the latency of hyperbaric oxygen-induced convulsion between the acetazolamide 200 mg/kg group and the NS control group, as well as between the acetazolamide 20 mg/kg group and the NS control group (P<0.01), whereas there was no significant difference between the NS group and the acetazolamide 2 mg/kg weight group (P>0.05). The latency of these groups were listed as follows: 9.78+/-1.94 min for 200 mg/kg body weight group, 10.92+/-1.68 min for 20 mg/kg body weight group, 24.32+/-4.33 min for 2 mg/kg body weight group and 22.02+/-4.32 min for NS control group. (2) there was no significant difference between all groups in the activity of GSH-PX, though it varied with the oxidation levels. In the cortex and hippocampus, the activity of GSH-PX boosted up at first, but with the progress of the oxidation it was impaired. In the striatum, the activity of GSH-PX increased stepwise with the aggravation of the oxidation. The MDA content in the cortex increased significantly in the group of 6 min with acetazolamide (P<0.01), as well as the group of 16 min with acetazolamide group both in cortex and hippocampus (P<0.01, P<0.05). The MDA content of all groups is correlated with the dose of acetazolamide and the exposure time. These results suggest that acetazolamide which dilates the brain arteriolar obviously shortens the latency of hyperbaric oxygen-induced convulsion, and that acetazolamide dilates the vessels and increases the supply of the oxygen breaking into the brain tissues and aggravates the oxidation. The hyperbaric oxygen-induced convulsion correlates closely with the oxidation injury.


Asunto(s)
Acetazolamida/farmacología , Oxigenoterapia Hiperbárica , Estrés Oxidativo , Convulsiones/fisiopatología , Animales , Encéfalo/patología , Masculino , Oxígeno , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Convulsiones/inducido químicamente , Vasodilatadores/farmacología
15.
Artículo en Chino | MEDLINE | ID: mdl-21166216

RESUMEN

AIM: To investigate the preventive effects of Panax notoginseng saponins (PNS) and Ginkgo biloba extracts (GbE) on acute oxygen toxicity and the possible mechanisms. METHODS: Mice were injected intraperitoneally with PNS and GbE for 5 days, then were exposed to 500 kPa hyperbaric oxygen (HBO) for 60 min, the convulsion latency, times and interval were observed. Moreover, reactive oxygen (RO) unit, MDA, NO, GSH levels and GSH-Px, CAT, MAO activities of mice brain were determined after they were exposed to HBO for 15 min. RESULTS: PNS and GbE could markedly prolong the convulsion latency and interval, reduce convulsion times, decrease contents of MDA and NO in mice brain, keep RO unit, GSH and GSH-Px at higher levels, but had no effects on CAT and MAO activities. CONCLUSION: PNS and GbE could effectively prevent acute oxygen toxicity, which were related to their antioxidant activities.


Asunto(s)
Ginkgo biloba , Oxígeno/envenenamiento , Panax notoginseng , Extractos Vegetales/farmacología , Saponinas/farmacología , Animales , Antioxidantes/farmacología , Buceo/efectos adversos , Oxigenoterapia Hiperbárica/efectos adversos , Masculino , Ratones , Fitoterapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA