Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Undersea Hyperb Med ; 50(2): 155-165, 2023.
Article in English | MEDLINE | ID: mdl-37302078

ABSTRACT

Background: This study sought to investigate therapeutic effects of hydrogen-rich saline (HRS) combined with hyperbaric oxygen (HBO2) in an experimental rat model of acute lung injury (ALI). Method: Forty male Sprague-Dawley rats were randomly divided into sham, LPS, LPS + HBO2, LPS + HRS, and LPS + HBO2 + HRS groups. After an intratracheal injection of LPS-induced ALI, the rats were given a single-agent HBO2 or HRS or HBO2 + HRS treatment. The treatments were continued for three days in this experimental rat model of ALI. At the end of experiment, the lung pathological, inflammatory factors, and cell apoptosis in the pulmonary tissue were detected by Tunel method and cell apoptosis rate was calculated accordingly. Results: In the groups treated with HBO2 + HRS, pulmonary pathological data, wet-dry weight ratio, and inflammatory factors of pulmonary tissues and alveolar lavage fluid were significantly superior to those of the sham group (p≺0.05). Cell apoptosis detection revealed that no single agent treatment of HRS or HBO2, or combination treatment, could alleviate all cell apoptosis. HRS combined with HBO2 treatment was superior to single treatment (p≺0.05). Conclusion: HRS or HBO2 single treatment could decrease inflammatory cytokines release in lung tissue, reduce the accumulation of oxidative products and alleviate apoptosis of pulmonary cells, then lead to positive therapeutic effects on ALI induced by LPS. Furthermore, HBO2 combined with HRS treatment presented a synergy effect on cell apoptosis decrease and a decline in inflammatory cytokine release and related inflammatory product generation, compared with a single treatment.


Subject(s)
Acute Lung Injury , Hyperbaric Oxygenation , Rats , Male , Animals , Rats, Sprague-Dawley , Lipopolysaccharides/adverse effects , Acute Lung Injury/chemically induced , Acute Lung Injury/prevention & control , Lung/pathology , Oxygen/adverse effects , Cytokines , Hydrogen/therapeutic use , Hydrogen/pharmacology
2.
J Transl Med ; 19(1): 288, 2021 07 03.
Article in English | MEDLINE | ID: mdl-34217324

ABSTRACT

BACKGROUND: This study aimed to establish and validate an easy-to-operate novel scoring system based on simple and readily available clinical indices for predicting the progression of chronic kidney disease (CKD). METHODS: We retrospectively evaluated 1045 eligible CKD patients from a publicly available database. Factors included in the model were determined by univariate and multiple Cox proportional hazard analyses based on the training set. RESULTS: Independent prognostic factors including etiology, hemoglobin level, creatinine level, proteinuria, and urinary protein/creatinine ratio were determined and contained in the model. The model showed good calibration and discrimination. The area under the curve (AUC) values generated to predict 1-, 2-, and 3-year progression-free survival in the training set were 0.947, 0.931, and 0.939, respectively. In the validation set, the model still revealed excellent calibration and discrimination, and the AUC values generated to predict 1-, 2-, and 3-year progression-free survival were 0.948, 0.933, and 0.915, respectively. In addition, decision curve analysis demonstrated that the model was clinically beneficial. Moreover, to visualize the prediction results, we established a web-based calculator ( https://ncutool.shinyapps.io/CKDprogression/ ). CONCLUSION: An easy-to-operate model based on five relevant factors was developed and validated as a conventional tool to assist doctors with clinical decision-making and personalized treatment.


Subject(s)
Renal Insufficiency, Chronic , Area Under Curve , Databases, Factual , Disease Progression , Humans , Internet , Retrospective Studies
3.
Undersea Hyperb Med ; 48(3): 239-245, 2021.
Article in English | MEDLINE | ID: mdl-34390628

ABSTRACT

Breathing less than 50 kPa of oxygen over time can lead to pulmonary oxygen toxicity (POT). Vital capacity (VC) as the sole parameter for POT has its limitations. In this study we try to find out the changes of acid-base status in a POT rat model. Fifty male rats were randomly divided into five groups, exposed to 230 kPa oxygen for three, six, nine and 12 hours, respectively. Rats exposed to air were used as controls. After exposure the mortality and behavior of rats were observed. Arterial blood samples were collected for acid-base status detection and wet-dry (W/D) ratios of lung tissues were tested. Results showed that the acid-base status in rats exposed to 230 kPa oxygen presented a dynamic change. The primary status was in the compensatory period when primary respiratory acidosis was mixed with compensated metabolic alkalosis. Then the status changed to decompensated alkalosis and developed to decompensated acidosis in the end. pH, PCO2, HCO3-, TCO2, and BE values had two phases: an increase and a later decrease with increasing oxygen exposure time, while PaO2 and lung W/D ratio showed continuously increasing trends with the extension of oxygen exposure time. Lung W/D ratio was significantly associated with PaO2 (r = 0.6385, p = 0.002), while other parameters did not show a significant correlation. It is concluded that acid-base status in POT rats presents a dynamic change: in the compensatory period first, then turns to decompensated alkalosis and ends up with decompensated acidosis status. Blood gas analysis is a useful method to monitor the development of POT.


Subject(s)
Acid-Base Imbalance/blood , Acidosis, Respiratory/metabolism , Alkalosis, Respiratory/metabolism , Hyperbaric Oxygenation/adverse effects , Oxygen/toxicity , Acid-Base Imbalance/etiology , Animals , Atmospheric Pressure , Bicarbonates/blood , Blood Chemical Analysis , Blood Gas Analysis , Carbon Dioxide/blood , Hyperbaric Oxygenation/methods , Lung/pathology , Male , Models, Animal , Organ Size , Partial Pressure , Random Allocation , Rats , Rats, Sprague-Dawley , Time Factors , Vital Capacity
4.
Undersea Hyperb Med ; 47(3): 445-453, 2020.
Article in English | MEDLINE | ID: mdl-32931671

ABSTRACT

The present study was designed to assess the stress responses to a simulation model of the undersea environment that is similar to some undersea working conditions such as submarine rescue, underwater salvage and underwater construction. Restraint, hyperbaric air and immersion were chosen to produce the simulation stress model in rats for four hours. Rats were randomized into five groups: control group, restraint (R) group, hyperbaric air (H) group, restraint plus hyperbaric air (RH) group, and restraint plus hyperbaric air plus immersion (RHI) group. The results showed that the responses to the simulation stress model of the undersea environment induced by R, H, RH and RHI involved the upregulated norepinephrine (NE), dopamine (DA) and 5-hydroxytryptamine (5-HT) of the central nervous system (CNS), upregulated adrenocorticotropic hormone (ACTH), corticosterone (CORT) and blood glucose of the neuroendocrine system, upregulated interleukin-1 (IL-1), interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α) of the immune system, and increased anxiety in rats. Compared with hyperbaric air, restraint tended to activate stronger stress responses. Conclusively, this work established a simulation stress model of the undersea environment induced by restraint, hyperbaric air and immersion. It further provided experimental data of such a model that showed significant activation of the CNS, neuroendocrine and immune systems and anxiety in rats. In this experiment we provided an experimental basis for undersea work such as working aboard a submarine.


Subject(s)
Anxiety/etiology , Central Nervous System/metabolism , Immune System/metabolism , Neurosecretory Systems/metabolism , Stress, Physiological/physiology , Adrenocorticotropic Hormone/metabolism , Animals , Blood Glucose/metabolism , Corticosterone/metabolism , Disease Models, Animal , Dopamine/metabolism , Elevated Plus Maze Test , Immersion , Interleukin-1/metabolism , Interleukin-6 , Male , Norepinephrine/metabolism , Open Field Test , Pressure , Random Allocation , Rats , Rats, Wistar , Restraint, Physical , Serotonin/metabolism , Simulation Training/methods , Stress, Psychological/physiopathology , Submarine Medicine , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
5.
FASEB J ; 32(9): 4670-4680, 2018 09.
Article in English | MEDLINE | ID: mdl-29601782

ABSTRACT

Exercise plays an important role in the prevention and treatment of chronic liver disease and associated metabolic disorders. A single bout of exercise induces tissue blood flow redistribution, which decreases splanchnic circulation and leads to physiologic hypoxia in the gastrointestinal system and liver. The transcription factor, hypoxia inducible factor-1α (HIF-1α), and its regulator, prolylhydroxylase 2 (PHD2), play pivotal roles in the response to oxygen flux by regulating downstream gene expression levels in the liver. We hypothesized that exercise increases the HIF-1α levels in the liver, and that the hepatic PHD2/HIF-1α axis is involved in postexercise restoration of systemic energy homeostasis. Through constant O2 consumption, CO2 production, food and water intake, and physical activity detection with metabolic chambers, we observed that one 30-min session of swimming exercise enhances systemic energy metabolism in mice. By using the noninvasive bioluminescence imaging ROSA26 oxygen-dependent domain Luc mouse model, we reveal that exercise increases in vivo HIFα levels in the liver. Intraperitoneal injections of the PHD inhibitor, dimethyloxalylglycine, mimicked exercise-induced HIFα increase, whereas the HIF-1α inhibitor, PX-478, blocked this effect. We next constructed liver-specific knockout (LKO) mouse models with albumin- Cre-mediated, hepatocyte-specific Hif1a and Phd2 deletion. Compared with their controls, Hif1a-LKO and Phd2-LKO mice exhibited distinct patterns of hepatic metabolism-related gene expression profiles. Moreover, Hif1a-LKO mice failed to restore systemic energy homeostasis after exercise. In conclusion, the current study demonstrates that a single bout of exercise disrupts systemic energy homeostasis, increasing the HIF-1α levels in the liver. These findings also provide evidence that the hepatic PHD2/HIF-1α axis is involved in postexercise systemic metabolic homeostasis.-Luo, B., Xiang, D., Wu, D., Liu, C., Fang, Y., Chen, P., Hu, Y.-P. Hepatic PHD2/HIF-1α axis is involved in postexercise systemic energy homeostasis.


Subject(s)
Homeostasis/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Liver/metabolism , Procollagen-Proline Dioxygenase/metabolism , Animals , Cell Line, Tumor , Gene Expression/genetics , Gene Expression Regulation/genetics , Mice, Transgenic , Oxygen/metabolism , Prolyl Hydroxylases/genetics , RNA, Messenger/genetics
6.
Plant J ; 92(6): 1157-1169, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29072877

ABSTRACT

Plants often develop the capacity to tolerate moderate and reversible environmental stresses, such as drought, and to re-establish normal development once the stress has been removed. An example of this phenomenon is provided by cut rose (Rosa hybrida) flowers, which experience typical reversible dehydration stresses during post-harvest handling after harvesting at the bud stages. The molecular mechanisms involved in rose flower dehydration tolerance are not known, however. Here, we characterized a dehydration- and abscisic acid (ABA)-induced ferritin gene (RhFer1). Dehydration-induced free ferrous iron (Fe2+ ) is preferentially sequestered by RhFer1 and not transported outside of the petal cells, to restrict oxidative stresses during dehydration. Free Fe2+ accumulation resulted in more serious oxidative stresses and the induction of genes encoding antioxidant enzyme in RhFer1-silenced petals, and poorer dehydration tolerance was observed compared with tobacco rattle virus (TRV) controls. We also determined that RhABF2, an AREB/ABF transcription factor involved in the ABA signaling pathway, can activate RhFer1 expression by directly binding to its promoter. The silencing of RhABF2 decreased dehydration tolerance and disrupted Fe homeostasis in rose petals during dehydration, as did the silencing of RhFer1. Although both RhFer1 and Fe transporter genes are induced during flower natural senescence in plants, the silencing of RhABF2 or RhFer1 accelerates the petal senescence processes. These results suggest that the regulatory module RhABF2/RhFer1 contributes to the maintenance of Fe levels and enhances dehydration tolerance through the action of RhFer1 locally sequestering free Fe2+ under dehydration conditions, and plays synergistic roles with transporter genes during flower senescence.


Subject(s)
Ferritins/metabolism , Iron/metabolism , Rosa/genetics , Transcription Factors/metabolism , Abscisic Acid/metabolism , Dehydration , Droughts , Ferritins/genetics , Flowers/cytology , Flowers/genetics , Flowers/physiology , Plant Growth Regulators/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism , Promoter Regions, Genetic/genetics , Rosa/cytology , Rosa/physiology , Stress, Physiological , Transcription Factors/genetics
7.
Am J Physiol Lung Cell Mol Physiol ; 314(2): L287-L297, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29074491

ABSTRACT

Statins, which are 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase competitive inhibitors, not only lower blood cholesterol but also exert pleiotropic and beneficial effects in various diseases. However, the effects of statins on acute lung injury (ALI) induced by hyperbaric oxygen (HBO) have not been investigated. The present study is the first to investigate the effects of simvastatin in ALI induced by HBO in 8- to 9-wk-old C57BL/6 mice exposed to 0.23 MPa [=2.3 atmosphere absolute (ATA)] hyperoxia (≥95% O2) for 6 h. Mice were either given simvastatin (20 mg·kg·-1·day-1) in saline or a saline vehicle for 3 days before oxygen exposure. Lung tissue, serum, and bronchoalveolar lavage fluid (BALF) were collected for analysis of proapoptotic proteins, low-density lipoprotein cholesterol (LDL-C) levels, and lung inflammation. Simvastatin treatment significantly reduced lung permeability, serum LDL-C levels, tissue apoptosis, and inflammation. However, simvastatin treatment had no effect on antioxidant enzyme activity, nicotinamide adenine dinucleotide phosphate oxidase 4 (NADPH4) expression, and Akt phosphorylation levels. Furthermore, we investigated the role of endothelial nitric oxide synthase (eNOS) in simvastatin protection through inhibiting eNOS activity with NG-nitro-l-arginine methyl ester (l-NAME; 20 mg/kg). Results showed that the beneficial effects of simvastatin on ALI induced by HBO (antiinflammatory, antiapoptotic, lipid lowering, and reduction in lung permeability) were reversed. These results showed that simvastatin curbs HBO-induced lung edema, permeability, inflammation, and apoptosis via upregulating eNOS expression and that simvastatin could be an effective therapy to treat prolonged HBO exposure.


Subject(s)
Acute Lung Injury/prevention & control , Anticholesteremic Agents/pharmacology , Gene Expression Regulation/drug effects , Hyperbaric Oxygenation/adverse effects , Nitric Oxide Synthase Type III/metabolism , Simvastatin/pharmacology , Acute Lung Injury/enzymology , Acute Lung Injury/etiology , Animals , Male , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/genetics , Transcriptional Activation
8.
Undersea Hyperb Med ; 45(3): 351-362, 2018.
Article in English | MEDLINE | ID: mdl-30028921

ABSTRACT

Nuclear factor kappa B (NF-κB) is the critical transcriptional factor in the pathogenesis of acute lung injury (ALI). NF-κB regulates the expression changes of inflammatory factors such as tumor necrosis factor alpha (TNF-α), interleukin-1ß (IL-1ß) and interleukin 6 (IL-6). In a previous study we showed that decompression sickness (DCS) caused by simulated unsafe fast buoyancy ascent escape (FBAE) could result in ALI, which was characterized by expression changes of inflammatory factors in rat lung tissue. The purpose of the present work was to study the roles of NF-κB and TNF-α in the process of DCS-induced rat lung injury caused by simulated unsafe FBAE. The research methods aimed to detect the rat lung tissue messenger ribonucleic acid (mRNA) and protein level variations of NF-κB, inhibitory ×B (I×B), TNF-α, IL-1ß, IL-6, IL-10 and IL-13 by using pretreatment of the NF-κB inhibitor pyrrolidine dithiocarbamate (PDTC) and TNF-α antibody (Ab). Our experimental results demonstrated that PDTC could improve the survival rate of the rats with DCS caused by unsafe FBAE more effectively than TNF-α Ab. However, the inhibition of TNF-α Ab on the nuclear translocated protein expression of NF-κB was more effective than PDTC. Both PDTC and TNF-α Ab can abrogate the increment of the rat lung tissue mRNA levels of TNF-α, IL-1ß, IL-6 and protein levels of NF-κB, TNF-α, IL-1ß effectively and increase the rat lung tissue content of I×B significantly. In conclusion, TNF-α-mediated NF-κB signaling may be one of the critical signaling pathways in the pathogenesis of DCS-induced rat lung injury caused by simulated unsafe FBAE. PDTC may ameliorate this type of injury partly through inhibiting the NF-κB pathway.


Subject(s)
Acute Lung Injury/metabolism , Antioxidants/pharmacology , Decompression Sickness/complications , Interleukins/metabolism , NF-kappa B/metabolism , Pyrrolidines/pharmacology , Thiocarbamates/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Acute Lung Injury/etiology , Acute Lung Injury/pathology , Animals , Interleukin-10/metabolism , Interleukin-13/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Lung/metabolism , Lung/pathology , Male , NF-kappa B/antagonists & inhibitors , RNA, Messenger/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Survival Rate , Tumor Necrosis Factor-alpha/antagonists & inhibitors
9.
J Asian Nat Prod Res ; 19(7): 645-650, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27240189

ABSTRACT

The pentacyclic triterpenoid corosolic acid was metabolized by Cunninghamella echinulata CGMCC 3.2000 to its C-24 aldehyde group metabolite and five other hydroxylated metabolites: madasiatic acid (2), 2α, 3ß, 7ß-trihydroxyurs-12-en-28-oic acid (3), 2α, 3ß, 15α-trihydroxyurs-12-en-28-oic acid (4), 2α, 3ß, 6ß, 7ß-tetrahydroxyurs-12-en-28-oic acid (5), 2α, 3ß, 7ß, 15α-tetrahydroxyurs-12-en-28-oic acid (6), and 2α, 3ß,7ß-trihydroxy-24-al-urs-12-en-28-oic acid (7); compounds 3, 5, and 7 were new compounds. The α-glucosidase inhibitory effects of the metabolites were also evaluated.


Subject(s)
Cunninghamella/metabolism , Glycoside Hydrolase Inhibitors/pharmacology , Hypoglycemic Agents/pharmacology , Triterpenes/pharmacology , Biotransformation , Diabetes Mellitus/drug therapy , Glycoside Hydrolase Inhibitors/chemistry , Hypoglycemic Agents/chemistry , Molecular Structure , Stereoisomerism , Triterpenes/chemistry , alpha-Glucosidases/drug effects , alpha-Glucosidases/metabolism
10.
Undersea Hyperb Med ; 42(2): 115-23, 2015.
Article in English | MEDLINE | ID: mdl-26094286

ABSTRACT

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


Subject(s)
Decompression Sickness/prevention & control , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Simvastatin/therapeutic use , Adiposity , Administration, Oral , Animals , Bronchoalveolar Lavage Fluid/chemistry , Decompression/methods , Decompression Sickness/epidemiology , Decompression Sickness/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Incidence , Inflammation/prevention & control , Lipids/blood , Lung/chemistry , Lung/pathology , Lung Injury/metabolism , Lung Injury/prevention & control , Male , Malondialdehyde/analysis , Organ Size , Pneumonia/prevention & control , Pulmonary Edema/diagnosis , Rats , Rats, Sprague-Dawley , Simvastatin/administration & dosage , Tumor Necrosis Factor-alpha/analysis
11.
Undersea Hyperb Med ; 42(1): 15-22, 2015.
Article in English | MEDLINE | ID: mdl-26094300

ABSTRACT

Fast buoyancy ascent escape is one of the major naval submarine escape maneuvers. Decompression sickness (DCS) is the major bottleneck to increase the depth of fast buoyancy ascent escape. Rapid decompression induces the release of inflammatory mediators and results in tissue inflammation cascades and a protective anti-inflammatory response. In our previous study, we found that DCS caused by simulated fast buoyancy ascent escape could induce acute lung injury (ALI) and the expression changes of the proinflammatory cytokines: tumor necrosis factor alpha (TNF-α), interleukin (IL)-1ß and IL-6 in rat lung tissue. In order to study the expression change characteristics of TNF-α, IL-1ß, IL-6, IL-10 and IL-13 in the rat lung of DCS caused by simulated fast buoyancy ascent escape, we detected the rat lung mRNA and protein levels of TNF-α, IL-1ß, IL-6, IL-10 and IL-13 at 0.5 hour after DCS caused by simulated fast buoyancy ascent escape (fast escape group), compared with the normal control group (control group) and diving DCS (decompression group). We observed that DCS caused by simulated fast buoyancy ascent escape could increase the mRNA levels of TNF-α, IL-1ß, IL-6, IL-10, and the protein levels of TNF-α, IL-10 in rat lung tissue. At the same time, we found that the protein level of IL-13 was also downregulated in rat lung tissue. TNF-α, IL-10 and IL-13 may be involved in the process of the rat lung injury of DCS caused by simulated fast buoyancy ascent escape. In conclusion, the expression changes of inflammatory factors in the rat lung of DCS caused by simulated fast buoyancy ascent escape were probably different from that in the rat lung of diving DCS, which indicated that the pathological mechanism of DCS caused by simulated fast buoyancy ascent escape might be different from that of diving DCS.


Subject(s)
Decompression Sickness/metabolism , Interleukins/metabolism , Lung/metabolism , Tumor Necrosis Factor-alpha/metabolism , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Decompression Sickness/etiology , Decompression Sickness/mortality , Decompression Sickness/pathology , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukin-13/genetics , Interleukin-13/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukins/genetics , Lung/pathology , Male , RNA, Messenger/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Submarine Medicine , Time Factors , Tumor Necrosis Factor-alpha/genetics
12.
Undersea Hyperb Med ; 42(1): 23-31, 2015.
Article in English | MEDLINE | ID: mdl-26094301

ABSTRACT

Fast buoyancy ascent escape is the general submarine escape manner adopted by the majority of naval forces all over the world. However, if hyperbaric exposure time exceeds the time limit, fast buoyancy ascent escape has a high risk to result in decompression sickness (DCS). Tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß) and IL-6 have been all implicated in the process of inflammation associated with acute lung injury (ALI). Our work demonstrated that DCS caused by simulated fast buoyancy ascent escape could induce ALß in the rat model. The purpose of the present work was to study the expression changes of TNF-α, IL-1ß and IL-6 in the rat lung affected by DCS caused by simulated fast buoyancy ascent escape. The lung tissue mRNA levels of TNF-α, Il-1ß and Il-6 were significantly increased at 0.5 hour after DCS caused by simulated fast buoyancy ascent escape. The lung contents of TNF-α, IL-1ß and IL-6 were at an expression peak at 0.5 hour, although showing no statistical difference when compared with the normal control group. In conclusion, the rat lung expression variations of TNF-α, IL-1ß and IL-6 are the most obvious at 0.5 hour within 24 hours after the lung injury by DCS caused by simulated fast buoyancy ascent escape.


Subject(s)
Decompression Sickness/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Lung/metabolism , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/metabolism , Acute Lung Injury/etiology , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Decompression Sickness/pathology , Interleukin-1beta/genetics , Interleukin-6/genetics , Lung/pathology , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Submarine Medicine , Time Factors , Tumor Necrosis Factor-alpha/genetics
13.
Exp Lung Res ; 40(3): 105-16, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24624894

ABSTRACT

Recent studies have demonstrated that peroxisome proliferator-activated receptor-beta/delta (PPAR-ß/δ) has a protective effect during lung injury induced by bleomycin and polymicrobial sepsis, but its function in pulmonary oxygen toxicity is unknown. In this study, we used GW0742, a PPAR-ß/δ agonist, and GSK0660, a PPAR-ß/δ antagonist, to test the role of PPAR-ß/δ in lung injury due to hyperbaric oxygen (HBO2) exposure. Lung injury was induced in rats by HBO2 exposure (2.3 ATA, 100%O2, 8 hours). Sixty male Sprague-Dawley rats were randomly divided into 6 groups: air+vehicle, air+GW0742, air+GSK0660, HBO2+vehicle, HBO2+GW0742, and HBO2+GSK0660. Rats were injected with vehicle or GW0742 (0.3 mg/kg, i.p.) or GSK0660 (1 mg/kg, i.p.) at 1 hour, 6 hours, and 12 hours before either air or oxygen exposure. Administration of GW0742 to rats exposed to HBO2 significantly reduced the observed lung injury, extravascular lung water, total protein levels in bronchoalveolar lavage fluid, and the levels of iNOS and nNOS in the lungs when compared to untreated rats exposed to HBO2. Treatment of rats with GSK0660 exacerbated lung injury and elevated the levels of nNOS and eNOS in the lungs. In addition, nNOS and eNOS knock-out mice were examined. The results indicated that after HBO2 exposure, the lung injury was obviously decreased in the nNOS(-/-)+GSK0660 mice compared to the wild-type +GSK0660 mice; furthermore, administration of GSK0660 significantly elevated the lung injury in the eNOS(-/-) mice. Collectively, these data indicate that PPAR-ß/δ activation can protect against pulmonary oxygen toxicity in the lungs of rats through changes in the expression of NOS.


Subject(s)
Acute Lung Injury/etiology , Nitric Oxide Synthase/metabolism , Oxygen/adverse effects , PPAR delta/metabolism , PPAR-beta/metabolism , Acute Lung Injury/metabolism , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR delta/agonists , PPAR delta/antagonists & inhibitors , PPAR-beta/agonists , PPAR-beta/antagonists & inhibitors , Random Allocation , Rats , Rats, Sprague-Dawley , Sulfones , Thiazoles , Thiophenes , Up-Regulation
14.
Undersea Hyperb Med ; 41(1): 1-7, 2014.
Article in English | MEDLINE | ID: mdl-24649711

ABSTRACT

OBJECTIVE: To observe MMP9 expression in rat lungs under different degrees of hyperbaric oxygen (HBO2) exposure and to observe the relationship of tissue damage and apoptosis rate in the lung tissue. METHODS: 40 Sprague Dawley (SD) rats were randomly divided into five groups: 250 kPa oxygen exposure for two-, four-, six- and eight-hour exposures and a "normal group," each n = 8. After hyperbaric oxygen treatment, the rats were euthanized immediately to collect lung tissue. We used HE staining to detect the pathological changes and immunohistochemistry to detect in situ expression of matrix metalloproteinase 9 (MMP9). Then we tested the expression level of caspase 3 in lung tissue by Western Blot. To understand the antioxidant capacity changes, we detected superoxide dismutase (SOD) activities and malondialdehyde (MDA) contents in lung tissue during HBO2 exposure. Finally, we exposed MMP9 knockout or wild-type mice under hyperbaric oxygen for six hours, and detected the pathological changes with HE staining. RESULT: Lung tissue damage changed gradually under different degrees of hyperbaric oxygen exposure, but eased in the six-hour group. However, MMP9 expression decreased initially and then was upregulated until it reached the peak after six hours of exposure; it then reduced significantly after an eight-hour exposure. Active caspase 3 reached the highest level after eight hours. While SOD activity was upregulated only after six hours of HBO2 exposure, MDA content increased after eight hours of exposure. CONCLUSIONS: MMP9 expression was elevated after exposure to hyperbaric oxygen. This is a compensatory mechanism of the body's antioxidant response by regulating the inflammatory response. This in turn helps to reduce the apoptosis rate and protects lung tissue from oxygen toxicity.


Subject(s)
Apoptosis , Hyperbaric Oxygenation , Lung/enzymology , Matrix Metalloproteinase 9/metabolism , Animals , Caspase 3/metabolism , Gene Knockout Techniques , Lung/metabolism , Lung/pathology , Male , Malondialdehyde/metabolism , Matrix Metalloproteinase 9/genetics , Random Allocation , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism , Time Factors , Up-Regulation
15.
Int J Biol Macromol ; 260(Pt 2): 129378, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38218262

ABSTRACT

Conductive hydrogels have been widely used as sensors owing to their tissue-like properties. However, the synthesis of conductive hydrogels with highly adjustable mechanical properties and multiple functions remains difficult to achieve yet highly needed. In this study, lignin hydrogel characterized by frost resistance, UV resistance, high conductivity, and highly adjustable mechanical properties without forming by-products was prepared through a rapid in-situ polymerization of acrylic acid/zinc chloride (AA/ZnCl2) aqueous solution containing lignin extract induced by the reversible quinone-catechol redox of the ZnCl2-lignin system at room temperature. Results revealed that the PAA/ZnCl2/lignin hydrogel exhibited mechanical properties with tensile stress (ranging from 0.08 to 3.28 MPa), adhesion to multiple surfaces (up to 62.05 J m-2), excellent frost resistance (-70-20 °C), UV resistance, and conductivity (0.967 S m-1), which further endow the hydrogel as potential strain and temperature sensor with wide monitor range (0-300 %), fatigue resistance, and quick response (70 ms for 150 % strain). This study proposed and developed a green, simple, economical, and efficient processing method for a hydrogel sensor in flexible wearable devices and man-machine interaction fields.


Subject(s)
Hydrogels , Lignin , Humans , Polymerization , Electric Conductivity , Quinones
16.
Neurosci Res ; 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38848903

ABSTRACT

Underwater exercise is becoming increasingly prevalent, during which brain function is necessary but is also at risk. However, no study has explored how prolonged exercise affect the brain in underwater environment. Previous studies have indicated that excessive exercise in common environment causes brain dysfunction but have failed to provide appropriate interventions. Numerous evidence has indicated the neuroprotective effect of hyperbaric oxygen preconditioning (HBO-PC). The objective of this study was to investigate the cognitive effect of prolonged underwater exercise (PUE) and to explore the potential neuroprotective effect of HBO-PC in underwater environment. Rats swimming for 3 h in a simulated hyperbaric chamber (2.0 ATA) was used to establish the PUE animal model and HBO-PC (2.5 ATA for 1, 3,5 times respectively) was administrated before PUE. The results demonstrated that PUE triggers anxiety-like behaviors, cognitive impairment accompanied by hippocampal dysfunction, microglia activation and neuroinflammation. Conversely, 3 HBO-PC rescued anxiety-like behaviors and cognitive impairment. Mechanistically, 3 HBO-PC reduced microglia activation and switched the activated microglia from a pro-inflammatory to neuroprotective phenotype. These findings illustrated that PUE induces anxiety-like behaviors and cognitive impairment and HBO-PC of proper frequency may provide an appropriate and less invasive intervention for protecting the brain in underwater exercise.

17.
Int J Biol Macromol ; 265(Pt 1): 130447, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38458280

ABSTRACT

The increased use and expansion of biomass applications offer a viable approach to diminish reliance on petroleum-derived resources and promote carbon neutrality. Cellulose, being the most abundant natural polymer on Earth, has garnered considerable attention. This study introduces a straightforward method to fabricate a cellulose-based multifunctional composite film designed for efficient light management, specifically featuring flame retardant and thermal-healing capabilities. The film incorporates a microfibrillated cellulose (MFC) matrix with functional components, namely benzoxazine resin (BR) and 2-hydroxyethyl methacrylate phosphate (HEMAP). Utilizing dynamic covalent crosslinking, the composite films exhibit satisfactory self-healing properties. The combined effects of BR and HEMAP contribute to the effective flame retardancy of the composite film. Furthermore, the resulting film shields ultraviolet and blue light, offering comfortable interior lighting by mitigating harsh light and extending light propagation. The film also demonstrates favorable water resistance and high tensile strength. The exceptional multifunctional properties, coupled with its safety and extended service life, position it as a potential optical management film for smart building materials.


Subject(s)
Cellulose , Flame Retardants , Polymers , Benzoxazines , Biomass
18.
Biomed Pharmacother ; 170: 115679, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38113632

ABSTRACT

Bacopaside I (BSI) is a natural compound that is difficult to absorb orally but has been shown to have antidepressant effects. The microbiota-gut-brain axis is involved in the development of depression through the peripheral nervous system, endocrine system, and immune system and may be a key factor in the effect of BSI. Therefore, this study aimed to investigate the potential mechanism of BSI in the treatment of depression via the microbiota-gut-brain axis and to validate it in a fecal microbiota transplantation model. The antidepressant effect of BSI was established in CUMS-induced mice using behavioral tests and measurement of changes in hypothalamicpituitaryadrenal (HPA) axis-related hormones. The improvement of stress-induced gut-brain axis damage by BSI was observed by histopathological sections and enzyme-linked immunosorbent assay (ELISA). 16 S rDNA sequencing analysis indicated that BSI could modulate the abundance of gut microbiota and increase the abundance of probiotic bacteria. We also observed an increase in short-chain fatty acids, particularly acetic acid. In addition, BSI could modulate the disruption of lipid metabolism induced by CUMS. Fecal microbiota transplantation further confirmed that disruption of the microbiota-gut-brain axis is closely associated with the development of depression, and that the microbiota regulated by BSI exerts a partial antidepressant effect. In conclusion, BSI exerts antidepressant effects by remodeling gut microbiota, specifically through the Lactobacillus and Streptococcus-acetic acid-neurotrophin signaling pathways. Furthermore, BSI can repair damage to the gut-brain axis, regulate HPA axis dysfunction, and maintain immune homeostasis.


Subject(s)
Gastrointestinal Microbiome , Mice , Animals , Depression/metabolism , Hypothalamo-Hypophyseal System , Pituitary-Adrenal System , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Acetates/pharmacology , Stress, Psychological/metabolism
19.
Undersea Hyperb Med ; 40(4): 313-8, 2013.
Article in English | MEDLINE | ID: mdl-23957201

ABSTRACT

Prolonged exposure to hyperbaric oxygen can cause pulmonary and nerve system toxicity. Although hyperbaric oxygen treatment has been used for a broad spectrum of ailments, the mechanisms of prolonged hyperbaric oxygen-induced lung injury are not fully understood. The purpose of the present work was to investigate the roles of ERK, p38, and caspase-3 in rat lung tissue exposed to hyperbaric oxygen at 2.3 atmospheres absolute (atm abs) for two, six and 10 hours. The results showed that the ERK and p38 were phosphorylated at two hours and reached a peak at six hours into exposure to hyperbaric oxygen. While the phosphorylation level of ERK decreased, p38 remained at a high level of activation at 10 hours. The activation of ERK and p38 was down-regulated when rats were exposed to normoxic hyperbaric nitrogen for 10 hours. However, caspase-3 was activated at six hours and 10 hours into exposure to hyperbaric oxygen. These results demonstrated different changes of activation of ERK and p38 during lung injury induced by prolonged exposure to hyperbaric oxygen. The time course changes of activated caspase-3 were similar to the process of p38 activation upon exposure to hyperbaric oxygen. In this way, activation of p38, not ERK, seems to be a mechanism associated with prolonged hyperbaric oxygen-induced lung injury.


Subject(s)
Caspase 3/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Hyperbaric Oxygenation/adverse effects , Lung Injury/enzymology , Oxygen/toxicity , p38 Mitogen-Activated Protein Kinases/metabolism , Analysis of Variance , Animals , Apoptosis , Enzyme Activation , Lung/pathology , Lung Injury/etiology , Lung Injury/pathology , Male , Nitrogen , Phosphorylation , Random Allocation , Rats , Rats, Sprague-Dawley , Time Factors
20.
Front Physiol ; 14: 1107782, 2023.
Article in English | MEDLINE | ID: mdl-36776974

ABSTRACT

Objective: If a damaged submarine cannot be rescued in time, it is necessary to carry out a submarine escape by free ascent. Decompression illness is the greatest threat to the safety of submariners. The maximum depth at which a safe escape can be carried out is unknown. This study intends to explore the maximum safe escape depth by observing the effects of simulated submarine escape at different depths on animal models. Methods: We evaluated pulmonary function indexes, blood gas values, blood cell counts, the myocardial enzyme spectrum, coagulation parameters, and proinflammatory cytokine levels in rats, electrocardiographic activity in rabbits after simulated 150-m, 200-m, 220-m, and 250-m submarine escape by free ascent. Results: An escape depth of 150 m did not cause significant changes in the indicators. An escape depth of >200 m led to pulmonary ventilation and gas diffusion dysfunction, hypoxemia, myocardial ischemia, and activation of the fibrinolytic and inflammatory systems. The magnitudes of the changes in the indicators were proportional to escape depth. Conclusion: An escape depth of 150 m in animal models is safe, whereas escape at > 200 m can be harmful.

SELECTION OF CITATIONS
SEARCH DETAIL