Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
BMC Womens Health ; 24(1): 333, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849811

ABSTRACT

BACKGROUND: Pregnant women faced great challenges and psychological and physiological changes of varying degrees during the omicron epidemic outbreak. It is important to recognize the potential impact of these challenges on the mental health of pregnant women and to provide appropriate resources and support to mitigate their effects. METHOD: By using the convenience sampling approach, a total of 401 pregnant women from two hospitals of different grades in two cities were included in the survey. The cross-sectional survey was conducted by basic characteristics, Generalized Anxiety Disorder (GAD-7), Patient Health Questionnaire (PHQ-9), Insomnia Severity Index (ISI) and self-made questionnaire. RESULTS: Insomnia affected 207 participants (51.6%), depression affected 160 participants (39.9%) and anxiety affected 151 participants (37.7%). Moreover, pregnant women in provincial capital city were more likely to experience anxiety, depression and insomnia than those in county-level city (P < 0.01). Pregnant women's anxiety, depression and insomnia were positively correlated with the severity of COVID-19 infection (P < 0.05). However, COVID-19 infection had no appreciable impact on maternal demand for termination of pregnancy and cesarean section (P > 0.05). CONCLUSION: Pregnant women frequently suffer from anxiety disorder, depression and insomnia as a result of the omicron pandemic in China. During this period, the community and medical professionals should provide more psychological counseling, conduct health education and offer virtual prenatal care to pregnant women (particularly in the provincial capital city).


Subject(s)
Anxiety , COVID-19 , Depression , Pregnant Women , Sleep Initiation and Maintenance Disorders , Humans , Female , COVID-19/epidemiology , COVID-19/psychology , China/epidemiology , Pregnancy , Adult , Cross-Sectional Studies , Depression/epidemiology , Depression/psychology , Anxiety/epidemiology , Anxiety/psychology , Pregnant Women/psychology , Sleep Initiation and Maintenance Disorders/epidemiology , Sleep Initiation and Maintenance Disorders/psychology , Surveys and Questionnaires , Young Adult , SARS-CoV-2 , Pregnancy Complications/epidemiology , Pregnancy Complications/psychology , Mental Health/statistics & numerical data
2.
Med Gas Res ; 13(4): 161-164, 2023.
Article in English | MEDLINE | ID: mdl-37077112

ABSTRACT

Methane has shown protective effects on a variety of diseases. Among these, neurological diseases have attracted much attention. However, there are many different indicators and application methods of methane in the treatment of neurological diseases. In this review, we summarize the indicators related to the protective effects of methane and evaluate the preparation and administration of methane. Thus, we hope to offer available indicators and effective ways to produce and administer methane in future research.


Subject(s)
Methane , Nervous System Diseases , Methane/pharmacology , Nervous System Diseases/drug therapy , Humans
3.
Med Gas Res ; 13(1): 23-28, 2023.
Article in English | MEDLINE | ID: mdl-35946219

ABSTRACT

Demyelination of the cerebral white matter is the most common pathological change after carbon monoxide (CO) poisoning. Notch signaling, the mechanism underlying the differentiation of astrocytes and oligodendrocytes, is critical to remyelination of the white matter after brain lesion. The purpose of this work was to determine the effects of hyperbaric oxygen (HBO) on Notch signaling pathway after CO poisoning for the explanation of the protective effects of HBO on CO-poisoning-related cerebral white matter demyelination. The male C57 BL/6 mice with severe CO poisoning were treated by HBO. And HBO therapy shortened the escape latency and improved the body mass after CO poisoning. HBO therapy also significantly suppressed protein and mRNA levels of Notch1 and Hes5 after CO poisoning. Our findings suggested that HBO could suppress the activation of Notch signaling pathway after CO poisoning, which is the mechanism underlying the neuroprotection of HBO on demyelination after severe CO poisoning.


Subject(s)
Carbon Monoxide Poisoning , Demyelinating Diseases , Hyperbaric Oxygenation , Animals , Carbon Monoxide Poisoning/therapy , Demyelinating Diseases/chemically induced , Demyelinating Diseases/therapy , Male , Mice , Oxygen , Signal Transduction
4.
Med Gas Res ; 13(2): 72-77, 2023.
Article in English | MEDLINE | ID: mdl-36204786

ABSTRACT

Diabetic peripheral neuropathy (DPN) is a complex disorder caused by long-standing diabetes. Oxidative stress was considered the critical creed in this DPN pathophysiology. Hydrogen has antioxidative effects on diabetes mellitus and related complications. However, there is still no concern on the beneficial effects of hydrogen in DPN. This paper aimed to evaluate the effects of exogenous hydrogen to reduce the severity of DPN in streptozotocin-induced diabetic rats. Compared with hydrogen-rich saline treatment, hydrogen inhalation significantly reduced blood glucose levels in diabetic rats in the 4th and 8th weeks. With regard to nerve function, hydrogen administration significantly attenuated the decrease in the velocity of motor nerve conduction in diabetic animals. In addition, hydrogen significantly attenuated oxidative stress by reducing the level of malondialdehyde, reactive oxygen species, and 8-hydroxy-2-deoxyguanosine and meaningfully enhanced the antioxidant capability by partially restoring the activities of superoxide dismutase. Further studies showed that hydrogen significantly upregulated the expression of nuclear factor erythroid-2-related factor 2 and downstream proteins such as catalase and hemeoxygenase-1 in the nerves of diabetic animals. Our paper showed that hydrogen exerts significant protective effects in DPN by downregulating oxidative stress via the pathway of nuclear factor erythroid-2-related factor 2, which suggests its potential value in clinical applications.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Neuropathies , Neuroprotective Agents , Animals , Rats , Antioxidants/metabolism , Antioxidants/pharmacology , Blood Glucose , Catalase/metabolism , Catalase/pharmacology , Catalase/therapeutic use , Deoxyguanosine/metabolism , Deoxyguanosine/pharmacology , Deoxyguanosine/therapeutic use , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetic Neuropathies/drug therapy , Diabetic Neuropathies/metabolism , Hydrogen , Malondialdehyde , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Oxidative Stress , Reactive Oxygen Species , Streptozocin , Superoxide Dismutase/metabolism , Superoxide Dismutase/pharmacology , Superoxide Dismutase/therapeutic use
7.
Transl Neurosci ; 9: 33-37, 2018.
Article in English | MEDLINE | ID: mdl-29992051

ABSTRACT

BACKGROUND: Present study evaluates the neuroprotective effect of ß-elemene alone and in combination with hyperbaric oxygen (HO) in traumatic brain injury (TBI). METHODOLOGY: TBI was induced by dropping a weight from a specific height. All the animals were separated in to five groups (n=20) like control group; TBI group; ß-elemene treated group which receives ß-elemene (100 mg/kg, i.p.) half an hour after the injury; HO group which receives hyperbaric oxygen therapy and ß-elemene + HO group which receives ß-elemene (100 mg/kg, i.p.) half an hour after the injury and hyperbaric oxygen therapy. Neurological function was assessed to evaluate the effect of ß-elemene in TBI rats. Thereafter level of inflammatory cytokines and expression of protein of inflammatory pathway was assessed in the brain tissues of TBI rats. In addition TUNEL assay was also done for the determination apoptosis in neuronal cells. RESULT: Data of the report reveals that ß-elemene alone and in combination with hyperbaric oxygen (HO) significantly decreases the neurological score Compared to TBI group. Moreover level of inflammatory cytokines and expression of LTR4 and casepase 3 significantly decrease and increase in the expression of IkB in ß-elemene alone and in combination with hyperbaric oxygen (HO) treated group compared to TBI group. Data of TUNEL assay also reveals that ß-elemene treated group shows significant decrease in the TUNEL positive cells and apoptosis index compared to TBI group. CONCLUSION: Thus present study concludes the neuroprotective effect of ß-elemene against TBI and it shows synergistic effect on TBI when treated with HO.

8.
Undersea Hyperb Med ; 44(3): 287-291, 2017.
Article in English | MEDLINE | ID: mdl-28779586

ABSTRACT

PURPOSE: Pulmonary edema following hyperbaric oxygen (HBO2) therapy is a rare clinical phenomenon. This case report describes such a patient - a 56-year-old woman who suffered from severe pulmonary edema after HBO2 therapy for carbon monoxide (CO) poisoning. CASE: Patient experienced ecphysesis and dyspnea suddenly after HBO2 therapy (100% oxygen at 0.25 MPa, for 60 minutes with a five-minute air break and decompression at 0.01 MPa/minute). Post therapy her heart rate (HR), blood pressure (BP), respiratory rate (RR) and oxygen saturation (SO2) were 140 bpm, 60/40 mmHg, 38 bpm and 84%, respectively. Diagnoses of acute pulmonary edema and shock were made. Various treatments including antishock, tracheal intubation, mechanical ventilation for respiratory support, a diuretic, dexamethasone, asthma relief, and acidosis correction were administered. Pulmonary computed tomography (CT) indicated significant pulmonary edema. Due to active treatment, the patient showed gradual improvement. Pulmonary CT re-examination showed pulmonary edema markedly improved. At the two-year follow-up, the patient reported no abnormal mental or neurological symptoms. CONCLUSION: Acute pulmonary edema is rare but can lead to serious side effects of HBO2 therapy in patients with severe acute CO poisoning. This complication must be must considered when administering HBO2 therapy to patients with severe CO poisoning.


Subject(s)
Carbon Monoxide Poisoning/therapy , Hyperbaric Oxygenation/adverse effects , Pulmonary Edema/etiology , Shock/etiology , Blood Pressure , Female , Heart Rate , Humans , Middle Aged , Oxygen/blood , Pulmonary Edema/diagnosis , Respiratory Rate , Shock/diagnosis
9.
Free Radic Res ; 51(6): 622-632, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28675985

ABSTRACT

Hyperoxic acute lung injury (HALI) is a major clinical problem for patients undergoing supplemental oxygen therapy. Currently in clinical settings there exist no effective means of prevention or treatment methods. Our previous study found that: hydrogen could reduce HALI, as well as oxidative stress. This research will further explore the mechanism underlying the protective effect of hydrogen on oxygen toxicity. Rats were randomly assigned into three experimental groups and were exposed in a oxygen chamber for 60 continuous hours: 100% balanced air (control); 100% oxygen (HALI); 100% oxygen with hydrogen treatment (HALI + HRS). We examined lung function by wet to dry ratio of lung, lung pleural effusion and cell apoptosis. We also detected endoplasmic reticulum stress (ERS) by examining the expression of CHOP, GRP78 and XBP1. We further investigated the role of Sirtuin 1 (SIRT1) in HALI, which contributes to cellular regulation including ERS, by examining its expression after hydrogen treatment with SIRT1 inhibitor. Hydrogen could significantly reduce HALI by reducing lung edema and apoptosis, inhibiting the elevating of ERS and increased SIRT1 expression. By inhibition of SIRT1 expression, the effect of hydrogen on prevention of HALI is significantly weakened, the inhibition of the ERS was also reversed. Our findings indicate that hydrogen could reduce HALI related ERS and the mechanism of hydrogen may be associated with upregulation of SIRT1, this study reveals the molecular mechanisms underlying the protective effect of hydrogen, which provides a new theoretical basis for clinical application of hydrogen.


Subject(s)
Acute Lung Injury/prevention & control , Hydrogen/pharmacology , Hyperoxia/prevention & control , Oxygen/adverse effects , Protective Agents/pharmacology , Sirtuin 1/genetics , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Animals , Apoptosis/drug effects , Carbazoles/pharmacology , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Enzyme Activation/drug effects , Gene Expression Regulation , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Histone Deacetylase Inhibitors/pharmacology , Hyperoxia/chemically induced , Hyperoxia/genetics , Hyperoxia/pathology , Male , Organ Size/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/metabolism , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism
10.
J Neurol Sci ; 369: 361-367, 2016 Oct 15.
Article in English | MEDLINE | ID: mdl-27653924

ABSTRACT

BACKGROUND: Methane has been reported to play a protective role in ischemia-reperfusion injury via anti-oxidation, anti-inflammatory and anti-apoptotic activities. This study was designed to determine the protective effects of methane-rich saline (MRS) on acute carbon monoxide (CO) poisoning. METHODS: A total of 36 male Sprague-Dawley rats were randomly divided into 3 groups: sham group, CO group and MRS group. Acute CO poisoning was induced by exposing rats to 1000ppm CO in air for 40min and then to 3000ppm CO for an additional 20min until they lost consciousness. MRS at 10ml/kg was intraperitoneally administered at 0h, 8h and 16h after CO exposure. Rats were sacrificed 24h after CO exposure. Brains were collected for Nissl staining. The cortex and hippocampus were separated for the detections of malondialdehyde (MDA), 3-nitrotyrosine (3-NT), 8-hydroxydeoxyguanosine (8-OHdG), tumor necrosis factor-α (TNF-α), interleukin1-ß (IL-1ß), interleukin-6 (IL-6) and superoxide dismutase (SOD) activities. RESULTS: The results showed that MRS treatment improved neuronal injury, reduced MDA, 3-NT and 8-OHdG, and increased SOD activity of the hippocampus and cortex compared with normal saline-treated rats. In addition, MRS reduced the expression of TNF-α and IL-1ß in the brain but had no effect on IL-6 expression. CONCLUSION: These findings suggest that MRS may protect the brain against acute CO poisoning-induced injury via its anti-oxidative and anti-inflammatory activities.


Subject(s)
Carbon Monoxide Poisoning/drug therapy , Methane/therapeutic use , Neuroprotective Agents/therapeutic use , Sodium Chloride/therapeutic use , 8-Hydroxy-2'-Deoxyguanosine , Analysis of Variance , Animals , Carbon Monoxide Poisoning/pathology , Carboxyhemoglobin/metabolism , Cell Count , Cytokines/metabolism , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Male , Malondialdehyde/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism , Time Factors , Tyrosine/analogs & derivatives , Tyrosine/metabolism
11.
Undersea Hyperb Med ; 43(1): 45-8, 2016.
Article in English | MEDLINE | ID: mdl-27000012

ABSTRACT

It has been known that the pathophysiology of carbon monoxide (CO) poisoning is related to hypoxia, the increased production of reactive oxygen species (ROS) and oxidative stress. Studies have shown that the novel, safe and effective free radical scavenger, hydrogen, has neuroprotective effects in both acute CO poisoning and delayed neuropsychological sequelae in CO poisoning. Orally administered lactulose, which may be used by some intestinal bacteria as a food source to produce endogenous hydrogen, can ameliorate oxidative stress. Based on the available findings, we hypothesize that oral administration of lactulose may be a novel therapy for acute CO poisoning via increasing intestinal hydrogen production.


Subject(s)
Carbon Monoxide Poisoning/therapy , Hydrogen/metabolism , Intestinal Mucosa/metabolism , Lactulose/administration & dosage , Administration, Oral , Humans , Oxidative Stress , Reactive Oxygen Species/metabolism
12.
Med Sci Monit ; 22: 284-8, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26812205

ABSTRACT

BACKGROUND: The aim of this study was to investigate the efficacy of hyperbaric oxygen in secondary brain injury after trauma and its mechanism in a rat model. MATERIAL/METHODS: A rat model of TBI was constructed using the modified Feeney's free-fall method, and 60 SD rats were randomly divided into three groups--the sham group, the untreated traumatic brain injury (TBI) group, and the hyperbaric oxygen-treated TBI group. The neurological function of the rats was evaluated 12 and 24 hours after TBI modeling; the expression levels of TLR4, IκB, p65, and cleaved caspase-3 in the peri-trauma cortex were determined by Western blot; levels of TNF-α, IL-6, and IL-1ß were determined by ELISA; and apoptosis of the neurons was evaluated by TUNEL assay 24 hours after TBI modeling. RESULTS: Hyperbaric oxygen therapy significantly inhibited the activation of the TLR4/NF-κB signaling pathway, reduced the expression of cleaved caspase-3, TNF-α, IL-6 and IL-1ß (P<0.05), reduced apoptosis of the neurons and improved the neurological function of the rats (P<0.05). CONCLUSIONS: Hyperbaric oxygen therapy protects the neurons after traumatic injury, possibly through inhibition of the TLR4/NF-κB signaling pathway.


Subject(s)
Brain Injuries/metabolism , Brain Injuries/therapy , Hyperbaric Oxygenation , NF-kappa B/metabolism , Signal Transduction , Toll-Like Receptor 4/metabolism , Animals , Apoptosis/drug effects , Brain Injuries/physiopathology , Caspase 3/metabolism , Cytokines/metabolism , I-kappa B Proteins/metabolism , In Situ Nick-End Labeling , Male , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Oxygen/pharmacology , Rats, Sprague-Dawley , Signal Transduction/drug effects , Transcription Factor RelA/metabolism
13.
Brain Res ; 1633: 62-72, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26711852

ABSTRACT

OBJECTIVE: Delayed neuropsychological sequelae (DNS) are the most common and serious effects of severe carbon monoxide (CO) poisoning, occurring in approximately half of all CO poisoning cases. Growing evidence suggests that oxidative stress and secondary reactions in delayed brain injury are crucial to CO toxicity, similar to ischaemia-reperfusion injury. Exogenous methane plays a protective role in ischaemia-reperfusion injury by affecting key events through anti-oxidant, anti-inflammatory, and anti-apoptosis actions. Our study aimed to explore the potential of exogenous methane to relieve the severity of DNS. METHODS: Thirty-six male Sprague-Dawley (SD) rats were divided into three groups of normal-, CO- and CO plus methane-treated rats. The rats in the latter two groups were exposed to 1000 ppm CO for 40 min and then to 3000 ppm CO for another 20 min. Following CO exposure, saline or methane saline (10 ml/kg) was intraperitoneally administered to rats in the CO group or the CO plus methane group, respectively. On the ninth day after CO exposure, Morris water maze testing, histological analysis, terminal deoxynucleotidyl transferase-mediated dUTP nick-end labelling (TUNEL) and immunohistochemical labelling were performed on 6 rats in each group. The remaining 6 rats in each group were used to detect oxidative damage markers, inflammatory cytokines and apoptosis proteins. RESULTS: Methane significantly improved CO-impaired pathological characteristics as well as learning and memory performance. In addition, methane significantly increased the superoxide dismutase (SOD) activity, lowered the CO-increased level of malondialdehyde (MDA) 3-nitrotyrosine (3-NT) and 8-hydroxy-2-deoxyguanosine (8-OHdG), inhibited levels of tumour necrosis factor-α (TNF-α), interleukin 1-ß (IL1-ß) and caspase-3 in the rat cerebral cortex and hippocampus but had no effect on IL-6 levels. CONCLUSION: The hippocampus was the main target of CO-induced alterations in the rat brain compared to the cerebral cortex. Methane treatment protected the rat brain from the harmful effects induced by CO exposure and improved the outcome of DNS through anti-oxidant, anti-inflammatory and anti-apoptosis activities.


Subject(s)
Brain/drug effects , Carbon Monoxide Poisoning/pathology , Methane/pharmacology , Neuroprotective Agents/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Blotting, Western , Brain/metabolism , Brain/pathology , Carbon Monoxide Poisoning/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , In Situ Nick-End Labeling , Male , Maze Learning/drug effects , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley
14.
Neurochem Res ; 41(4): 770-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26537817

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is the most frequent adult-onset motor neuron disease, and accumulating evidence indicates that oxidative mechanisms contribute to ALS pathology, but classical antioxidants have not performed well in clinical trials. The aim of this work was to investigate the effect of treatment with hydrogen molecule on the development of disease in mutant SOD1 G93A transgenic mouse model of ALS. Treatment of mutant SOD1 G93A mice with hydrogen-rich saline (HRS, i.p.) significantly delayed disease onset and prolonged survival, and attenuated loss of motor neurons and suppressed microglial and glial activation. Treatment of mutant SOD1 G93A mice with HRS inhibited the release of mitochondrial apoptogenic factors and the subsequent activation of downstream caspase-3. Furthermore, treatment of mutant SOD1 G93A mice with HRS reduced levels of protein carbonyl and 3-nitrotyrosine, and suppressed formation of reactive oxygen species (ROS), peroxynitrite, and malondialdehyde. Treatment of mutant SOD1 G93A mice with HRS preserved mitochondrial function, marked by restored activities of Complex I and IV, reduced mitochondrial ROS formation and enhanced mitochondrial adenosine triphosphate synthesis. In conclusion, hydrogen molecule may be neuroprotective against ALS, possibly through abating oxidative and nitrosative stress and preserving mitochondrial function.


Subject(s)
Amyotrophic Lateral Sclerosis/prevention & control , Hydrogen/therapeutic use , Neuroprotective Agents/therapeutic use , Sodium Chloride/therapeutic use , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Apoptosis , Humans , Mice, Transgenic , Mitochondria/physiology , Motor Neurons/pathology , Neuroglia/pathology , Oxidative Stress , Spinal Cord/pathology , Superoxide Dismutase/genetics , Superoxide Dismutase-1
15.
Shock ; 44(2): 181-7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26009821

ABSTRACT

Hepatic ischemia/reperfusion (I/R) injury, which occurs in various diseases, introduces severe tissue damage and liver dysfunction. However, no promising therapies for such a significant condition currently exist. Methane has been suggested to exert a protective effect against intestinal I/R injury. In this study, we introduced methane to treat hepatic I/R injury to show its promising protective effect. Also, intraperitoneal injection with methane-rich saline, which could have potential clinical applications, was applied as a new method. Partial liver warm ischemia was applied in Sprague-Dawley rats for 60 min followed by succedent reperfusion. In the test for effective dosage, methane-rich saline was administrated intraperitoneally to the rats at doses of 1, 5, 20, or 40 mL/kg at onset of reperfusion. In the test for protective effect, rats received methane-rich saline intraperitoneally at a dose of 10 mL/kg before the initiation of reperfusion. We found that methane-rich saline significantly decreased serum alanine aminotransferase, aspartate aminotransferase activity, and the occurrence of necrosis. Moreover, methane-rich saline reduced the amount of caspase-3 and the number of apoptotic cells. In addition, methane-rich saline increased the level of superoxide dismutase and decreased the level of malondialdehyde and 8-hydroxyguanosine. Furthermore, research indicated that methane-rich saline markedly decreased gene expression and content of tumor necrosis factor-α and interleukin-6. Also, reduced CD68-positive cells showed decreased inflammatory cells in the liver. Our results suggest that methane protects the liver against I/R injury through antiapoptotic, antioxidative, and anti-inflammatory actions.


Subject(s)
Anti-Inflammatory Agents/chemistry , Antioxidants/chemistry , Apoptosis , Liver/pathology , Methane/chemistry , Reperfusion Injury/drug therapy , Alanine Transaminase/blood , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Aspartate Aminotransferases/blood , Caspase 3/metabolism , Dose-Response Relationship, Drug , Guanosine/analogs & derivatives , Guanosine/metabolism , Liver/metabolism , Male , Malondialdehyde/metabolism , Methane/administration & dosage , Necrosis/pathology , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism , Warm Ischemia
16.
Undersea Hyperb Med ; 40(4): 329-37, 2013.
Article in English | MEDLINE | ID: mdl-23957203

ABSTRACT

The aim of the present study was to examine the effects of simulated heliox diving at high altitudes on divers' blood cells, liver functions and renal functions. In this experiment, four divers lived for nine consecutive days in a dual-function high-low pressure chamber, which simulated air pressure at an altitude of 3,000 meters and at a 30-meter depth; an altitude of 4,000 meters and 30-meter depth; and at an altitude of 5,200 meters and 30 meters and 50 meters in depth. Total time underwater was 60 minutes. The subjects breathed heliox (with oxygen at 40% and helium at 60%) during the simulated 30-meter dive from zero altitude to 30 meters and while remaining underwater; they breathed air while ascending from 30 meters to 18. They breathed heliox (with oxygen at 26.7% and helium at 73.3%) in the simulated dive from zero altitude to 50 meters underwater, in remaining underwater and in ascending from 50 meters to 29; air while ascending from 29 meters to 18. Pure oxygen was breathed while ascending from 18 meters to the surface; then air. Results indicated: (1) the correlating indices of routine blood, liver and renal functions, and urine routine were all within normal reference ranges; and (2) the indices tested at other periods of time were not significantly different (p > 0.05) from the results at zero-meter level and 3,000-meter level. The study suggests that the heliox diving processes at different high altitudes simulated in this experiment have no significant impact upon divers' blood routine, liver functions and renal functions.


Subject(s)
Altitude , Blood Cell Count/methods , Diving/physiology , Helium/administration & dosage , Kidney/physiology , Liver/physiology , Oxygen/administration & dosage , Adult , Atmosphere Exposure Chambers , Biomarkers/blood , Biomarkers/urine , Decompression/methods , Diving/psychology , Humans , Hydrogen-Ion Concentration , Male , Middle Aged , Urine/chemistry , Urine/cytology
17.
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
18.
Undersea Hyperb Med ; 38(5): 335-43, 2011.
Article in English | MEDLINE | ID: mdl-22013760

ABSTRACT

The spinal cord is one of the most commonly affected sites in decompression sickness (DCS). Alternative methods have long been sought to protect against DCS spinal cord dysfunction, especially when hyperbaric treatment is unavailable. Use of perfluorocarbon (PFC) emulsion with or without oxygen breathing has shown survival benefits in DCS animal models. The effectiveness of intravenous PFC emulsion with oxygen breathing on spinal cord function was studied. Somatosensory-evoked potentials (SSEPs) and histologic examination were chosen to serve as measures. After fast decompression (203 kPa/minute) from 709 kPa (for 60 minutes), male Sprague-Dawley rats randomly received: 1) air and saline; 2) oxygen (O2) and saline; 3) O2 and PFC emulsion. The incidence and average number of abnormal SSEP waves in survival animals that received O2 and PFC emulsion were significantly reduced (P < 0.05). Foci of demyelination, necrosis and round non-staining defects in white matter regions of the spinal cord could be found in severe DCS rats. We concluded that administration of PFC emulsion combined with oxygen breathing was beneficial for DCS spinal conductive dysfunction in rats.


Subject(s)
Decompression Sickness/complications , Fluorocarbons/administration & dosage , Oxygen Inhalation Therapy/methods , Spinal Cord Injuries/therapy , Animals , Combined Modality Therapy/methods , Demyelinating Diseases/pathology , Emulsions , Evoked Potentials, Somatosensory/physiology , Infusions, Intravenous/methods , Leukoencephalopathies/pathology , Male , Necrosis , Random Allocation , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/etiology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology
19.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...