Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Neurotox Res ; 36(1): 193-203, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30927242

ABSTRACT

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.


Subject(s)
Adenosine/metabolism , Cerebral Cortex/metabolism , Nitric Oxide/metabolism , Oxygen/toxicity , Adenosine/administration & dosage , Adenosine Kinase/metabolism , Animals , Indazoles/administration & dosage , Lung/pathology , Male , Nitric Oxide Synthase Type I/antagonists & inhibitors , Rats, Sprague-Dawley
2.
Med Gas Res ; 7(4): 236-240, 2017.
Article in English | MEDLINE | ID: mdl-29497483

ABSTRACT

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.

3.
Brain Res ; 1635: 180-9, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26806404

ABSTRACT

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.


Subject(s)
Adenosine/metabolism , Astrocytes/metabolism , Brain/metabolism , Oxygen/toxicity , 5'-Nucleotidase/metabolism , Adenosine/administration & dosage , Animals , Brain/drug effects , Infusions, Intraventricular , Male , Rats , Rats, Sprague-Dawley
4.
Neuroreport ; 27(2): 73-9, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26619231

ABSTRACT

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.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Glutamic Acid/metabolism , Oxygen/toxicity , Air Pressure , Animals , Ceftriaxone/administration & dosage , Excitatory Amino Acid Transporter 2/agonists , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Glutamate-Ammonia Ligase/metabolism , Kainic Acid/administration & dosage , Kainic Acid/analogs & derivatives , Male , Rats , Rats, Sprague-Dawley
5.
CNS Neurosci Ther ; 21(12): 943-52, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26511680

ABSTRACT

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.


Subject(s)
Cell Hypoxia/drug effects , Glucose/deficiency , Hippocampus/drug effects , Ischemic Attack, Transient/drug therapy , Neurons/drug effects , Neuroprotective Agents/pharmacology , Sesquiterpenes/pharmacology , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/physiology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Hypoxia/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Hippocampus/metabolism , Hippocampus/pathology , Infarction, Middle Cerebral Artery , Ischemic Attack, Transient/metabolism , Ischemic Attack, Transient/pathology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , NF-kappa B/metabolism , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Random Allocation , Rats, Sprague-Dawley , Sesquiterpenes/chemistry
6.
Int Immunopharmacol ; 24(2): 182-190, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25523460

ABSTRACT

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.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Flavonoids/administration & dosage , Macrophages, Peritoneal/drug effects , Medicine, Chinese Traditional/methods , Shock, Septic/drug therapy , Animals , Cells, Cultured , Immunosuppression Therapy , Inflammation Mediators/metabolism , Interleukin-6/metabolism , Macrophages, Peritoneal/immunology , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Nitric Oxide/metabolism , Phosphorylation/drug effects , Rubus/immunology , Shock, Septic/immunology , Shock, Septic/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism
8.
CNS Neurol Disord Drug Targets ; 13(5): 874-84, 2014.
Article in English | MEDLINE | ID: mdl-24040794

ABSTRACT

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.


Subject(s)
Ischemic Attack, Transient/prevention & control , NF-kappa B/metabolism , Neuroprotective Agents/therapeutic use , Sesquiterpenes/therapeutic use , Animals , Animals, Newborn , Apoptosis/drug effects , Brain Infarction/etiology , Brain Infarction/prevention & control , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Hippocampus/cytology , In Situ Nick-End Labeling , Ischemic Attack, Transient/complications , Male , Nervous System Diseases/drug therapy , Nervous System Diseases/etiology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Sesquiterpenes/chemistry , Sesquiterpenes/pharmacology
9.
Exp Biol Med (Maywood) ; 238(1): 12-22, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23479759

ABSTRACT

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.


Subject(s)
Decompression Sickness/pathology , Decompression Sickness/physiopathology , HSP70 Heat-Shock Proteins/metabolism , Hyperbaric Oxygenation , Animals , Blotting, Western , Gene Expression Profiling , Lung/chemistry , Lung/pathology , Male , Rats , Rats, Sprague-Dawley , Severity of Illness Index , Spinal Cord/chemistry , Spinal Cord/pathology
11.
Phytomedicine ; 19(8-9): 677-81, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22483554

ABSTRACT

Polydatin is one of the most common encountered stilbenes of nature and a key component of the Chinese herb Polygonum cuspidatum. This study is to investigate the effects of polydatin on learning and memory impairments induced by chronic cerebral hypoperfusion in rats, as well as the potential mechanism. Both common carotid arteries and both vertebral arteries occlusion (four-vessel occlusion, 4-VO) induced severe cognitive deficits tested by water maze task, along with oxidative stress in hippocampus. Oral administration of polydatin for 30 days markedly attenuated cognitive deficits compared with the control (p < 0.05). Biochemical determination revealed that polydatin decreased the production of malondialdehyde (MDA) and significantly increased the activities of superoxide dismutase (SOD) and catalase (CAT). Additionally, polydatin effectively alleviated the injuries of cultured neurons induced by oxygen-glucose deprivation (OGD). These results suggest that polydatin exhibit therapeutic potential for vascular dementia, which is most likely related, at least in part, to its anti-oxidant activity and the direct protection of neurons.


Subject(s)
Dementia, Vascular/drug therapy , Glucosides/pharmacology , Learning/drug effects , Memory Disorders/drug therapy , Neuroprotective Agents/pharmacology , Stilbenes/pharmacology , Animals , Catalase/metabolism , Cells, Cultured , Disease Models, Animal , Drugs, Chinese Herbal , Fallopia japonica/chemistry , Hippocampus/drug effects , Hippocampus/metabolism , Male , Malondialdehyde/metabolism , Maze Learning/drug effects , Neurons/drug effects , Neurons/metabolism , Protective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism
12.
Aviat Space Environ Med ; 82(6): 604-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21702310

ABSTRACT

INTRODUCTION: Hydrogen (H2) has been reported to be effective in the treatment of oxidative injury, which plays an important role in the process of decompression sickness (DCS). This study was designed to test whether H2-rich saline (saline saturated with molecular hydrogen) protected rats against DCS. METHODS: Models of DCS were induced in male Sprague-Dawley rats weighing 300-310 g. H2-rich (0.86 mmol x L(-1)) saline was administered intraperitoneally (10 ml x kg(-1)) at 24 h, 12 h, immediately before compression, and right after fast decompression. RESULTS: H2-rich saline significantly decreased the incidence of DCS from 67.57 to 35.14% and partially counteracted the increases in the total concentration of protein in the bronchoalveolar lavage from 0.33 +/- 0.05 to 0.14 +/- 0.01 mg x ml(-1) (mean +/- SD; P < 0.05), myeloperoxidase activity from 0.86 +/- 0.16 to 0.44 +/- 0.13 U/g, levels of malondialdehyde (MDA) from 0.80 +/- 0.10 to 0.48 +/- 0.05 nmol x mg(-1), 8-hydroxydeoxyguanosine from 253.7 +/- 9.3 to 191.2 +/- 4.8 pg x mg(-1) in the lungs, and MDA level from 1.77 +/- 0.20 to 0.87 +/- 0.23 nmol x mg(-1) in the spinal cord in rat DCS models. The histopathology results also showed that H2-rich saline ameliorated DCS injuries. DISCUSSION: It is concluded that H2-rich saline may have a protective effect against DCS, possibly due to its antioxidant action.


Subject(s)
Decompression Sickness/prevention & control , Hydrogen/pharmacology , Sodium Chloride/pharmacology , 8-Hydroxy-2'-Deoxyguanosine , Analysis of Variance , Animals , Bronchoalveolar Lavage Fluid/chemistry , Cerebral Cortex/metabolism , Chi-Square Distribution , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Hydrogen/administration & dosage , Injections, Intraperitoneal , Lung/metabolism , Male , Malondialdehyde/metabolism , Oxidative Stress , Peroxidase/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Sodium Chloride/administration & dosage , Spinal Cord/metabolism
13.
Undersea Hyperb Med ; 37(3): 173-80, 2010.
Article in English | MEDLINE | ID: mdl-20568547

ABSTRACT

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.


Subject(s)
Decompression Sickness/prevention & control , Hyperbaric Oxygenation/methods , Nitric Oxide/metabolism , Animals , Brain/metabolism , Decompression Sickness/metabolism , Enzyme Inhibitors/administration & dosage , Male , Motor Activity/physiology , NG-Nitroarginine Methyl Ester/administration & dosage , Nitric Oxide/analysis , Nitrogen/administration & dosage , Oxygen/administration & dosage , Rats , Rats, Sprague-Dawley , Spinal Cord/metabolism , Time Factors
14.
Planta Med ; 75(3): 230-5, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19085813

ABSTRACT

Four novel bakkenolides - bakkenolide-Ia ( 1), bakkenolide-IIa ( 2), bakkenolide-IIIa ( 3) and bakkenolide-IVa ( 4) - were isolated from the extract of the rhizome of Petasites tricholobus. The structures were characterized by using NMR ( (1)H, (13)C, (1)H- (1)H COSY, HMQC, HMBC, and NOESY) and mass spectrometry. The neuroprotective activity of the compounds 1 - 4 was assayed with primary cultured neurons exposed to oxygen-glucose deprivation and oxidative insults. Antioxidant activity of the bakkenolides was evaluated by cell-free bioassays. The IN VITRO assay results showed that all these compounds exhibited significant neuroprotective and antioxidant activities. To our knowledge, this is the first report on the neuroprotective and antioxidant activities of bakkenolides.


Subject(s)
4-Butyrolactone/analogs & derivatives , 4-Butyrolactone/pharmacology , Antioxidants/pharmacology , Neuroprotective Agents/pharmacology , Petasites/chemistry , Spiro Compounds/pharmacology , 4-Butyrolactone/chemistry , 4-Butyrolactone/isolation & purification , Animals , Antioxidants/chemistry , Antioxidants/isolation & purification , Glucose/metabolism , L-Lactate Dehydrogenase/metabolism , Molecular Structure , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/chemistry , Neuroprotective Agents/isolation & purification , Oxygen/metabolism , Rats , Rhizome , Spiro Compounds/chemistry , Spiro Compounds/isolation & purification
15.
Article in Chinese | MEDLINE | ID: mdl-21166216

ABSTRACT

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.


Subject(s)
Ginkgo biloba , Oxygen/poisoning , Panax notoginseng , Plant Extracts/pharmacology , Saponins/pharmacology , Animals , Antioxidants/pharmacology , Diving/adverse effects , Hyperbaric Oxygenation/adverse effects , Male , Mice , Phytotherapy
SELECTION OF CITATIONS
SEARCH DETAIL
...