Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Clin Sci (Lond) ; 135(14): 1733-1750, 2021 07 30.
Article in English | MEDLINE | ID: mdl-34236078

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a worldwide chronic inflammatory lung disease, and influenza A virus (IAV) infection is a common cause of acute exacerbations of COPD (AECOPD). Therefore, targeting viral infections represents a promising strategy to prevent the occurrence and development of inflammatory flare ups in AECOPD. Jianpiyifei II (JPYFII) is a traditional herbal medicine used in China to treat patients with COPD, and its clinical indications are not well understood. However, investigation of the anti-inflammatory effects and underlying mechanism using an animal model of smoking have been reported in a previous study by our group. In addition, some included herbs, such as Radix astragali and Radix aupleuri, were reported to exhibit antiviral effects. Therefore, the aim of the present study was to investigate whether JPYFII formulation relieved acute inflammation by clearing the IAV in a mouse model that was exposed to cigarette smoke experimentally. JPYFII formulation treatment during smoke exposure and IAV infection significantly reduced the number of cells observed in bronchoalveolar lavage fluid (BALF), expression of proinflammatory cytokines, chemokines, superoxide production, and viral load in IAV-infected and smoke-exposed mice. However, JPYFII formulation treatment during smoke exposure alone did not reduce the number of cells in BALF or the expression of Il-6, Tnf-a, and Il-1ß. The results demonstrated that JPYFII formulation exerted an antiviral effect and reduced the exacerbation of lung inflammation in cigarette smoke (CS)-exposed mice infected with IAV. Our results suggested that JPYFII formulation could potentially be used to treat patients with AECOPD associated with IAV infection.


Subject(s)
Herbal Medicine , Influenza A virus/pathogenicity , Pneumonia/therapy , Pulmonary Disease, Chronic Obstructive/therapy , Animals , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Influenza, Human/complications , Lung/metabolism , Mice, Inbred BALB C , Pulmonary Disease, Chronic Obstructive/metabolism , Smoke/adverse effects , Smoking/adverse effects
2.
Clin Sci (Lond) ; 133(4): 551-564, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30733313

ABSTRACT

Chronic Obstructive Pulmonary Disease (COPD) is a major incurable global health burden and will become the third largest cause of death in the world by 2030. It is well established that an exaggerated inflammatory and oxidative stress response to cigarette smoke (CS) leads to, emphysema, small airway fibrosis, mucus hypersecretion, and progressive airflow limitation. Current treatments have limited efficacy in inhibiting chronic inflammation and consequently do not reverse the pathology that initiates and drives the long-term progression of disease. In particular, there are no effective therapeutics that target neutrophilic inflammation in COPD, which is known to cause tissue damage by degranulation of a suite of proteolytic enzymes including neutrophil elastase (NE). Matrine, an alkaloid compound extracted from Sophora flavescens Ait, has well known anti-inflammatory activity. Therefore, the aim of the present study was to investigate whether matrine could inhibit CS-induced lung inflammation in mice. Matrine significantly reduced CS-induced bronchoalveolar lavage fluid (BALF) neutrophilia and NE activity in mice. The reduction in BALF neutrophils in CS-exposed mice by matrine was not due to reductions in pro-neutrophil cytokines/chemokines, but rather matrine's ability to cause apoptosis of neutrophils, which we demonstrated ex vivo Thus, our data suggest that matrine has anti-inflammatory actions that could be of therapeutic potential in treating CS-induced lung inflammation observed in COPD.


Subject(s)
Alkaloids/pharmacology , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Cigarette Smoking , Lung Injury/prevention & control , Lung/drug effects , Neutrophils/drug effects , Quinolizines/pharmacology , Animals , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Leukocyte Elastase/metabolism , Lung/immunology , Lung/metabolism , Lung/pathology , Lung Injury/immunology , Lung Injury/metabolism , Lung Injury/pathology , Male , Mice, Inbred BALB C , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology , Peroxidase/metabolism , Smoke , Matrines
3.
Cell Physiol Biochem ; 47(5): 1800-1808, 2018.
Article in English | MEDLINE | ID: mdl-29953977

ABSTRACT

BACKGROUND/AIMS: Reactive oxygen species (ROS) are considered fundamental in various physiological/pathophysiological processes and prevention/treatment measures such as hyperbaric oxygen (HBO) therapy. In this study, the origination of ROS in human umbilical vein endothelial cells was investigated under basal and HBO conditions. METHODS: Whole cell or mitochondria-targeted fluorescent probes were applied to mark superoxide anion (O2-), and the ROS produced from mitochondrial respiratory chain (MRC), NADPH oxidase (NOX) and xanthine oxidase (XO) were identified by flow cytometry, confocal imaging and microplate fluorometry with or without specific inhibitors. An algorithm was established to calculate ROS proportion of each source. RESULTS: HBO increased ROS to about 2.14-2.44 fold in mitochondria and 1.32-1.42 fold in whole cell. Then ROS levels were significantly decreased by MRC inhibition about 30% and 16%, respectively. NOX or XO inhibition did not affect HBO-induced ROS generation. Based on these data, it could be further estimated that mitochondrial ROS accounted for 32%-39% of basal whole-cell ROS including 3% from MRC complex II, and NOX accounted for at least 24%-29%. Following HBO treatment, almost all increased ROS originated from mitochondria, and MRC complex II contributed at least 45%-60%. CONCLUSION: This study provided a simple but effective method to estimate the origination of intracellular ROS and found that MRC were the main source of HBO-induced ROS generation.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , Hyperbaric Oxygenation , Oxygen/metabolism , Reactive Oxygen Species/metabolism , Electron Transport , Humans , Mitochondria/metabolism , Oxygen/pharmacology , Reactive Oxygen Species/analysis
4.
Cell Physiol Biochem ; 46(3): 965-974, 2018.
Article in English | MEDLINE | ID: mdl-29669319

ABSTRACT

BACKGROUND/AIMS: Intravascular bubbles can exert pleiotropic detrimental effects, partly by inducing endothelial microparticles (EMPs) production, which play critical roles in cell communication and vascular inflammation cascades. However, the underlying mechanisms remain unclear. This study aimed to delineate the possible mechanisms involving bubble-induced EMPs formation. METHODS: Human umbilical vein endothelial cells (HUVECs) were contacted by bubbles and EMPs level in supernatant were quantified by flow cytometry. Cytoplasmic calcium (Ca2+) was measured by the Ca2+ binding dyes Fluo-3 AM and flippase activity was assessed by translocation rate of fluorescent phosphatidylserine (PS) analogue NBD-PS. Protein levels of phospho-myosin light chain (MLC, a Rho kinase substrate) and phospho-extracellular signal-regulated kinase 1 or 2 (ERK1/2) were determined by western blotting. The score of actin colocalization was assessed by phalloidin-FITC using an immunofluorescent microscopy. RESULTS: EMPs level markedly increased after bubble stimulus. Cytoplasmic calcium (Ca2+) significantly elevated (P< 0.05), flippase activity decreased (P< 0.05), protein levels of phospho- MLC and phospho- ERK1/2 significantly increased (P< 0.05, P < 0.05), and the score of actin colocalization markedly reduced (P< 0.05) in bubble-stimulated HUVECs. All the above changes except the increase in phospho-ERK1/2 can be reversed by Ca2+ channel blocker LaCl3 (P< 0.05). Additionally, MLC phosphorylation was significantly inhibited and actin colocalization markedly increased by Rho kinase inhibitor pretreatment and more importantly, bubble-induced EMPs markedly decreased. CONCLUSIONS: These results demonstrate that bubble stimulates EMPs formation by cytoplasmic Ca2+ elevation and subsequently activating Rho kinase pathway and cytoskeleton reorganization. Simultaneously, cytoplasmic Ca2+ inhibits the flippase activity and subsequently increases phosphatidylserine exposure, which also contributes to EMPs formation.


Subject(s)
Calcium/metabolism , Cell-Derived Microparticles/metabolism , rho-Associated Kinases/metabolism , Actins/metabolism , Aniline Compounds/chemistry , Calcium/chemistry , Cytoplasm/metabolism , Flavonoids , Human Umbilical Vein Endothelial Cells , Humans , Lanthanum/pharmacology , Microscopy, Fluorescence , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Myosin Light Chains/metabolism , Phosphatidylserines/metabolism , Phosphorylation/drug effects , Xanthenes/chemistry , rho-Associated Kinases/antagonists & inhibitors
5.
Cell Mol Neurobiol ; 36(1): 121-30, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26271288

ABSTRACT

Elevated ß-amyloid (Aß) is a hallmark of Alzheimer's disease (AD). Recent evidence has suggested that the receptor of advanced glycation end products (RAGE) is a key target for Aß-induced perturbation in AD, and blockade of RAGE significantly alleviates synaptic injury. Our previous study has suggested that ß-asarone could reduce neuronal apoptosis and improve memory deficits in ß-amyloid precursor protein and presenilin-1 (APP/PS1) double transgenic AD-model mice. In the present study, we evaluated the effects of ß-asarone on amyloidosis in APP/PS1 mice. We found that the survival of neurons of APP/PS1 mice was improved by ß-asarone, meanwhile, ß-asarone decreased Aß deposition and down-regulated Aß1-42 levels in cortex and hippocampus of APP/PS1 mice brain. Interestingly, the level of RAGE was also significantly down-regulated by ß-asarone. Our findings suggest that ß-asarone might be effective for the treatment of AD, and the decreasing effects of ß-asarone on Aß might associate with its down-regulation of RAGE.


Subject(s)
Alzheimer Disease/drug therapy , Amyloidosis/drug therapy , Anisoles/therapeutic use , Down-Regulation/drug effects , Receptor for Advanced Glycation End Products/metabolism , Allylbenzene Derivatives , Alzheimer Disease/complications , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloidosis/complications , Amyloidosis/pathology , Animals , Anisoles/administration & dosage , Anisoles/pharmacology , Cell Survival/drug effects , Cerebral Cortex/pathology , Disease Models, Animal , Hippocampus/pathology , Mice, Transgenic , Neurons/drug effects , Neurons/pathology , Plaque, Amyloid/complications , Plaque, Amyloid/drug therapy , Plaque, Amyloid/pathology , Presenilin-1/metabolism
6.
Undersea Hyperb Med ; 43(4): 421-426, 2016.
Article in English | MEDLINE | ID: mdl-28763171

ABSTRACT

OBJECTIVE: To explore the possible effects of rapid decompression on the activity and function of vascular endothelial cells in vitro. METHODS: Human umbilical vein endothelial cell (HUVEC) cultures were exposed at 7 atmospheres absolute (atm abs) air for two hours before decompression. Two decompression profiles were used at the rate of 30 atm abs min-1 (rapid decompression) or 1 atm abs min-1 (normal decompression). Three hours after decompression, cell activity was detected by cell counting kit-8 (CCK-8) assay and lactate dehydrogenase (LDH) activity assay; cell permeability was measured by electrical resistance determinations. Twelve hours after decompression, cell apoptosis was detected by flow cytometry with Annexin V FITC/PI double staining. RESULTS: There was no significant statistical difference between rapid and normal decompression groups in all the determined parameters (P=0.59, 0.87, 0.86 and 0.81, respectively). CONCLUSIONS: HUVECs endure rapid decompression well from 7 atm abs at the rate of 30 atm abs min-1, or the current determinations are not sensitive enough to reveal the possible injuries. Further research with more sensitive indexes is warranted to reveal the possible effects and mechanisms.


Subject(s)
Apoptosis , Atmospheric Pressure , Cell Survival , Decompression/methods , Human Umbilical Vein Endothelial Cells/physiology , L-Lactate Dehydrogenase/metabolism , Analysis of Variance , Cell Count/instrumentation , Cell Membrane Permeability , Decompression/adverse effects , Flow Cytometry , Human Umbilical Vein Endothelial Cells/metabolism , Humans , In Vitro Techniques , Time Factors
7.
Phytomedicine ; 126: 155273, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38342020

ABSTRACT

BACKGROUND: Chronic obstructive pulmonary disease (COPD) is defined by persistent airway and lung inflammation, excessive mucus production, remodeling of the airways, and damage to the alveolar tissue. Based on clinical experience, it has been observed that Jianpiyifei II (JPYF II) granules exhibit a significant therapeutic impact on individuals suffering from stable COPD. Nevertheless, the complete understanding of JPYF II's potential mode of action against COPD remains to be further clarified. PURPOSE: To further investigate the underlying mechanism of JPYF II for treating COPD and clarify the role of the IL-17 pathway in the treatment. METHODS: A variety of databases were utilized to acquire JPYF II's bioactive components, as well as related targets of JPYF II and COPD. Cytoscape was utilized to establish multiple interaction networks for the purpose of topological analyses and core-target screening. The Metascape was utilized to identify the function of target genes and crucial signaling pathways. To evaluate the interactions between bioactive ingredients and central target proteins, molecular docking simulations were conducted. Following that, a sequence of experiments was conducted both in the laboratory and in living organisms, which included analyzing the cell counts in bronchoalveolar lavage fluid (BALF), examining lung tissue for histopathological changes, conducting immunohistochemistry, RT‒qPCR, ELISA, and Western blotting. RESULTS: In JPYF II, 88 bioactive ingredients were predicted to have a total of 342 targets. After conducting Venn analysis, it was discovered that 284 potential targets of JPYF II were linked to the provision of defensive benefits against COPD. The PPI network yielded a total of twenty-four core targets. The findings from the analysis of enrichment and gene‒pathway network suggested that JPYF II targeted Hsp90, MAPKs, ERK, AP-1, TNF-α, IL-6, COX-2, CXCL8, and MMP-9 as crucial elements for COPD treatment through the IL-17 pathway. Additionally, JPYF II might modulate MAPK signaling pathways and the downstream transcription factor AP-1 via IL-17 regulation. According to the findings from molecular docking, it was observed that the 24 core target proteins exhibited robust binding affinities towards the top 10 bioactive compounds. Furthermore, the treatment of COPD through the regulation of MAPKs in the IL-17 pathway was significantly influenced by flavonoids and sterols found in JPYF II. In vitro, these observations were further confirmed. In vivo results demonstrated that JPYF II reduced inflammatory cell infiltration in pulmonary tissues and the quantity of inflammatory cells in BALF obtained from LPS- and CS-stimulated mice. Moreover, the administration of JPYF II resulted in the inhibition of IL-17 mRNA and protein levels, phosphorylation levels of MAPK proteins, and expression of phosphorylated AP-1 proteins. It also suppressed the expression of downstream effector genes and proteins associated with the IL-17/MAPK/AP-1 signaling axis in lung tissues and BALF. CONCLUSION: This research reveals that JPYF II improves COPD by controlling the IL-17/MAPK/AP-1 signaling axis within the IL-17 pathway for the first time. These findings offer potential approaches for the creation of novel medications that specifically target IL-17 and proteins involved in the IL-17 pathway to address COPD.


Subject(s)
Drugs, Chinese Herbal , Pulmonary Disease, Chronic Obstructive , Animals , Mice , Molecular Docking Simulation , Interleukin-17 , Network Pharmacology , Transcription Factor AP-1 , Pulmonary Disease, Chronic Obstructive/drug therapy , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use
8.
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.

9.
PLoS One ; 18(3): e0282700, 2023.
Article in English | MEDLINE | ID: mdl-36917582

ABSTRACT

The main objective of the present study was to determine metabolic profile changes in the brains of rats after simulated heliox saturated diving (HSD) to 400 meters of sea water compared to the blank controls. Alterations in the polar metabolome in the rat brain due to HSD were investigated in cortex, hippocampus, and striatum tissue samples by applying an NMR-based metabolomic approach coupled with biochemical detection in the cortex. The reduction in glutathione and taurine levels may hypothetically boost antioxidant defenses during saturation diving, which was also proven by the increased malondialdehyde level, the decreased superoxide dismutase, and the decreased glutathione peroxidase in the cortex. The concomitant decrease in aerobic metabolic pathways and anaerobic metabolic pathways comprised downregulated energy metabolism, which was also proven by the biochemical quantification of the metabolic enzymes Na-K ATPase and LDH in cerebral cortex tissue. The significant metabolic abnormalities of amino acid neurotransmitters, such as GABA, glycine, and aspartate, decreased aromatic amino acids, including tyrosine and phenylalanine, both of which are involved in the metabolism of dopamine and noradrenaline, which are downregulated in the cortex. Particularly, a decline in the level of N-acetyl aspartate is associated with neuronal damage. In summary, hyperbaric decompression of a 400 msw HSD affected the brain metabolome in a rat model, potentially including a broad range of disturbing amino acid homeostasis, metabolites related to oxidative stress and energy metabolism, and destabilizing neurotransmitter components. These disturbances may contribute to the neurochemical and neurological phenotypes of HSD.


Subject(s)
Diving , Rats , Animals , Oxidative Stress/physiology , Amino Acids/metabolism , Energy Metabolism , Superoxide Dismutase/metabolism , Cerebral Cortex/metabolism , Metabolome , Neurotransmitter Agents/metabolism
10.
Phytomedicine ; 100: 154057, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35349835

ABSTRACT

BACKGROUND: Complementary and alternative therapy is widely used to treat chronic obstructive pulmonary disease (COPD). A Chinese herbal medicine, JianPiYiFei (JPYF) II granules, have been shown to improve COPD patients' quality of life, however long-term effectiveness has not been examined. PURPOSE: To investigate whether long-term treatment with JPYF II granules is effective and safe for patients with stable, moderate to very severe COPD. STUDY DESIGN AND METHODS: A multicentre, randomised, double-blinded, placebo-controlled trial was conducted. Eligible participants from six hospitals were randomly assigned 1:1 to receive either JPYF II granules or placebo for 52 weeks. The primary outcome was the change in St. George's Respiratory Questionnaire (SGRQ) score during treatment. Secondary outcomes included the frequency of acute exacerbations during treatment, COPD Assessment Test (CAT), 6-minute walking test (6MWT), lung function, body mass index, airflow obstruction, dyspnoea, exercise capacity (BODE) index, and peripheral capillary oxygen saturation (SpO2) at the end of treatment. RESULTS: A total of 276 patients (138 in each group) were included in the analysis. JPYF II granules led to a significantly greater reduction in SGRQ score (-7.33 points, 95% CI -10.59 to -4.07; p < 0.0001) which reflects improved quality of life. JPYF II granules improved CAT (-3.49 points, 95% CI -5.12 to -1.86; p < 0.0001) and 6MWT (45.61 metres, 95% CI 20.26 to 70.95; p = 0.0005), compared with placebo. Acute exacerbations were less frequent with JPYF II granules than with placebo (0.87 vs. 1.34 events per patient; p = 0.0043). There were no significant differences between the groups in lung function, BODE index and SpO2. JPYF II granules were well tolerated and no significant adverse effects were noted. CONCLUSIONS: Long-term treatment with JPYF II granules is effective in moderate to very severe COPD, improving quality of life and exercise capacity, decreasing the risk of acute exacerbation, and relieving symptoms.


Subject(s)
Pulmonary Disease, Chronic Obstructive , Quality of Life , Disease Progression , Humans , Pulmonary Disease, Chronic Obstructive/drug therapy
11.
J Ethnopharmacol ; 278: 114312, 2021 Oct 05.
Article in English | MEDLINE | ID: mdl-34107330

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: The genus Erycibe belongs to the Convolvulaceae family that contains approximately 70 species mainly distributed from tropical and subtropical Asia to north of Australia. Several Erycibe species are traditionally used in folk medicine for the treatment of various ailments, including rheumatic arthralgia, primary glaucoma, hepatopathies, and infectious and malignant diseases. AIM OF THE REVIEW: This review aims to summarize comprehensive and updated information on traditional medicinal uses, phytochemistry, pharmacology, and toxicology of Erycibe species to provide a reference for the further research and application of the Erycibe genus. MATERIALS AND METHODS: The scientific and extensive literatures between 1975 and 2020 were systematically gathered from scientific databases such as SciFinder Scholar, Science Direct, Web of Science, PubMed, Google Scholar, Scopus, Springer Link and China National Knowledge Infrastructure (CNKI), as well as Chinese herbal classic books, PhD and MSc theses, and several official websites. RESULTS: Erycibe species have been used for the treatment of various rheumatoid diseases, glaucoma, a variety of hepatic diseases, infectious diseases and various malignancies in the traditional and local medicine. Since the 1970s, 153 compounds, including coumarins, quinic acid derivatives, flavonoids, alkaloids, lignans, and others have been isolated from five species of the Erycibe genus. Pharmacological studies have shown that these extracts and compounds from the Erycibe genus have extensive activities consistent with the traditional and local applications, such as anti-glaucoma, anti-arthritic, hepatoprotective and anti-cancer activities, as well as anti-inflammatory, anti-respiratory syncytial virus (RSV), and neuroprotective properties. CONCLUSIONS: Although there are extensive data on the genus Erycibe, certain specific gaps still exist. For herbal preparations containing Erycibe species, clinical toxicological investigation is required for the safety of these herbal preparation therapies, as well as further investigations on pharmacokinetics and bioavailability for guideline for clinical application. Furthermore, more detailed pharmacological, toxicological and clinical researches are needed to assess the alternatives to Erycibe species. Systematic and comprehensive pre-clinical studies are similarly required to estimate the possibility of extracts and compounds from the genus Erycibe with bioactivity developing into new drugs.


Subject(s)
Convolvulaceae/chemistry , Convolvulaceae/classification , Ethnopharmacology , Phytochemicals , Phytotherapy , Plants, Medicinal/chemistry , Humans
12.
J Appl Physiol (1985) ; 130(2): 400-407, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33270509

ABSTRACT

Decompression sickness (DCS) is a systemic pathophysiological process featured by bubble load. Lung dysfunction plays a harmful effect on off-gassing, which contributes to bubble load and subsequent DCS occurrence. This study aimed to investigate the effects of pulmonary surfactant on DCS as it possesses multiple advantages on the lung. Rats were divided into three groups: the normal (n = 10), the surfactant (n = 36), and the saline (n = 36) group. Animals in surfactant or saline group were administered aerosol surfactant or saline 12 h before a stimulated diving, respectively. Signs of DCS were recorded and bubble load was detected. The contents of phospholipid and surfactant protein A (SPA), protein, IL-1 and IL-6 in bronchoalveolar lavage fluid (BALF), and lung wet/dry (W/D) ratio were determined. Serum levels of IL-6, ICAM-1, E-selectin, GSH, and GSSG were detected. In surfactant-treated rats, the morbidity and mortality of DCS markedly decreased (P < 0.01 and P < 0.05, respectively). Survival time prolonged and the latency to DCS dramatically delayed (P < 0.01). More importantly, bubble load markedly decreased (P < 0.01). The increases of protein, IL-1 and IL-6 in BALF, and lung W/D ratio were alleviated. Restoration of total phospholipid and SPA in BALF and ICAM-1 and E-selectin in serum was observed. The inflammation and oxidation were attenuated (P < 0.01). In conclusion, prediving administrating exogenous surfactant by aerosolization is an efficient, simple, and safe method for DCS prevention in rats.NEW & NOTEWORTHY This is the first study exploring the effects of aerosol surfactant on DCS prevention and it was proven to be an efficient and simple method. The role of surfactant in facilitating off-gassing was thought to be the critical mechanism in bubble degrading and subsequent DCS prevention.


Subject(s)
Decompression Sickness , Diving , Pulmonary Surfactants , Animals , Lung , Rats , Rats, Sprague-Dawley
13.
Front Pharmacol ; 12: 603863, 2021.
Article in English | MEDLINE | ID: mdl-34887746

ABSTRACT

Aims: The occurrence of vascular permeability pulmonary edema in acute lung injury (ALI) is related to the imbalance of alveolar fluid transport. Regulating the active transport of alveolar fluid by aquaporins (AQPs), epithelial sodium channels (ENaCs), and Na+-K+-ATPase can effectively reduce the edema fluid in the alveolar cavity and protect against ALI. We evaluated the therapeutic effects of total flavonoids, extracted from Nervilia fordii (TFENF), and investigated its potential mechanisms of alveolar fluid transport in a rat ALI model. Materials and methods: A model of lipopolysaccharide (LPS, 5 mg/kg)-induced ALI was established in Sprague-Dawley (SD) rats through the arteriae dorsalis penis. SD rats were divided into six groups, including the vehicle, LPS model, TFENF (6 mg/kg, 12 mg/kg, 24 mg/kg), and dexamethasone group (DEX group, 5 mg/kg). The wet-to-dry (W/D) lung weight ratio, oxygenation index, and histopathological observation were used to evaluate the therapeutic effect of TFENF. The mRNA expression of AQPs, ENaCs, and pro-inflammatory cytokines was determined using real-time polymerase chain reaction, whereas protein expression was determined using immunohistochemistry. The Na + -K + -ATPase activity was assessed using enzyme-linked immunosorbent assay. Results: LPS significantly stimulated the production of inflammatory mediators including tumor necrosis factor (TNF)-α and interleukin (IL)-1ß, and disrupted the water transport balance in the alveolar cavity by inhibiting AQPs/ENaCs/Na + -K + -ATPase. Pretreatment with TFENF reduced the pathological damage and W/D ratio of the lungs and ameliorated the arterial blood oxygen partial pressure (PaO2) and oxygenation index. TFENF further decreased the mRNA level of TNF-α and IL-1ß; increased the expression of AQP-1, AQP-5, αENaC, and ßENaC; and increased Na + -K + -ATPase activity. Moreover, the regulation of AQPs, ßENaC, and Na + -K + -ATPase and the inhibition of TNF-α and IL-1ß by TFENF were found to be dose dependent. Conclusion: TFENF protects against LPS-induced ALI, at least in part, through the suppression of inflammatory cytokines and regulation of the active transport capacity of AQPs/ENaCs/Na + -K + -ATPase. These findings suggest the therapeutic potential of TFENF as phytomedicine to treat inflammation and pulmonary edema in ALI.

14.
Front Pharmacol ; 11: 581, 2020.
Article in English | MEDLINE | ID: mdl-32425799

ABSTRACT

Jianpiyifei II granules (JPYF II), a herbal formula, are used for the treatment of chronic obstructive pulmonary disease (COPD) in Guangdong Provincial Hospital of Chinese Medicine. The protective effects of JPYF II against bronchial epithelial cell apoptosis in mice exposed to cigarette smoke (CS) and apoptosis of human bronchial epithelial cell lines (BEAS-2B and 16-HBE) stimulated with cigarette smoke extract (CSE) were investigated. Mice were exposed to CS generated from four cigarettes/day for 30 days and administered a dose of JPYF II (0.75, 1.5, and 3 g/kg/d) from the 3rd week of CS exposure. In mice exposed to CS, JPYF II significantly inhibited CS-induced apoptosis and overexpression of endoplasmic reticulum (ER) stress-related markers in bronchial epithelial cells of the lung tissues. In CSE-stimulated BEAS-2B and 16-HBE cells, JPYF II attenuated apoptosis and cell cycle arrest in the G0/G1 phase. Mechanistically, CSE initially induced intracellular reactive oxygen species (ROS) production, which then triggered ER stress, leading to the release of Ca2+ from ER inositol trisphosphate receptor (IP3R)-mediated stores and finally cell death. Treatment with JPYF II resulted in a significant reduction in CSE-induced apoptosis through interruption of the ROS-ER stress-Ca2+ signaling pathway. Therefore, the results of this study have revealed the underlying mechanism of action of JPYF II in the treatment of COPD.

15.
Chin J Integr Med ; 26(11): 857-866, 2020 Nov.
Article in English | MEDLINE | ID: mdl-31776962

ABSTRACT

OBJECTIVE: To assess the efficacy and safety of Chinese medicine injection (CMI) for treating acute lung injury/acute respiratory distress syndrome (ALI/ARDS). METHODS: Randomized controlled trials (RCTs) were identified by searching 3 English databases and 4 Chinese databases from their inceptions until February 2019. The Cochrane Handbook was used to evaluate risk of bias in the included studies. Data analysis was conducted using RevMan 5.3.3 software. RESULTS: A total of 19 eligible RCTs involving 1,334 participants was included in this systematic review and meta-analysis. The main meta-analysis showed that CMI combined with conventional therapy (CT) was more effective than CT alone in reducing the acute physiology and chronic health evaluation (APACHE) H score [mean difference (MD): -1.74 points, 95% confidence interval (CI): -2.77 to -0.71, I2=0] and increasing the total effective rate [relative risk (RR): 1.35, 95% CI: 1.17 to 1.56, I2=37%]. Compared with CT, CMI combined with CT showed improvements in the arterial partial pressure of oxygen (PaO2, MD: 9.25 mm Hg, 95% CI: 0.87 to 17.63, I2=98%) and oxygenation index [arterial partial pressure of oxygen (PaO2)/fraction of inspired oxygen (FiO2), MD: 50.75 mm Hg, 95% CI: 35.18 to 66.31, I2=94%]. CMI plus CT was superior to CT in reducing the systemic inflammatory response syndrome (SIRS) score (MD: -0.84 points, 95% CI: -1.26 to -0.42, I2=65%), length of hospital stay (MD: -4.22 days, 95% CI: -6.49 to -1.95, I2=92%), and duration of mechanical ventilation (MD: -2.94 days, 95% CI: -4.68 to -1.21, I2=89%). Only 1 study reported adverse events. CONCLUSIONS: CMI as an adjuvant therapy showed great potential benefits for the treatment of ALI/ARDS. However, we could not make a definite conclusion due to low quality of included studies and uncertain security. Future studies should focus on improving research design, especially in blindness and placebo. The reporting of adverse events was also needed.


Subject(s)
Acute Lung Injury/drug therapy , Medicine, Chinese Traditional/methods , Respiratory Distress Syndrome/drug therapy , Humans , Injections , Randomized Controlled Trials as Topic
16.
Biomed Pharmacother ; 120: 109427, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31648165

ABSTRACT

Gastric cancer is recognized as one of the most common cancer. In-depth research of gastric precancerous lesions (GPL) plays an important role in preventing the occurrence of gastric cancer. Meanwhile, traditional treatment provides a novel sight in the prevention of occurrence and development of gastric cancer. The current study was designed to assess the effects of therapy with Weipixiao (WPX) decoction on N-methyl-N'-nitro-N-nitrosoguanidine (MNNG)-induced GPL rats and the underlying molecular mechanisms. After 10-weeks treatment, all rats were sacrificed. Histopathological changes of gastric tissue were assessed via hematoxylin-eosin (HE) and High-iron diamine-Alcian blue-Periodic acid-Schiff (HID-AB-PAS) staining. To be fully evidenced, RT-qPCR, Western blot and immunohistochemistry were used to detect the expressions of LDHA, CD147, HIF-1α, MCT4, PI3K, AKT, mTOR and miRNA-34a, which were crucial factors for evaluating GPL in the aspect of glycolysis pathogenesis. According to the results of HE and HID-AB-PAS staining, it could be confirmed that MNNG-induced GPL rats were obviously reversed by WPX decoction. Additionally, the increased gene levels of LDHA, CD147, MCT4, PI3K, AKT, mTOR and HIF-1α in model group were down-regulated by WPX decoction, while miRNA-34a expression was decreased and up-regulated by WPX decoction. The significantly increased protein levels of LDHA, CD147, MCT4, PI3K, AKT, mTOR and HIF-1α induced by MNNG were attenuated in rats treated with WPX decoction. In brief, the findings of this study imply that abnormal glycolysis in MNNG-induced GPL rats was relieved by WPX decoction via regulation of the expressions of LDHA, CD147, HIF-1α, MCT4, PI3K, AKT, mTOR and miRNA-34a.


Subject(s)
Anticarcinogenic Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Glycolysis/drug effects , Methylnitronitrosoguanidine , Precancerous Conditions/prevention & control , Stomach Neoplasms/prevention & control , Stomach/drug effects , Animals , Cytoprotection , Disease Models, Animal , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Male , Precancerous Conditions/chemically induced , Precancerous Conditions/enzymology , Precancerous Conditions/pathology , Rats, Sprague-Dawley , Signal Transduction , Stomach/enzymology , Stomach/pathology , Stomach Neoplasms/chemically induced , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology
17.
Article in English | MEDLINE | ID: mdl-31073318

ABSTRACT

Cordyceps sinensis (CS) is a complementary medicine used for Chronic Obstructive Pulmonary Disease (COPD) of Global Initiative for Chronic Obstructive Lung Disease (GOLD) stages 2-3. Many randomized controlled trials have been conducted to evaluate the effect of CS alone or in combination with other herbs on stable COPD. To provide a synthesis of the evidence, we searched nine major electronic databases for randomized controlled trials on CS published before 21st December 2016. Fifteen interventional studies, including 1,238 participants, met the inclusion criteria. Meta-analysis showed that both CS preparations and CS formulae showed the potential benefits in lung function, exercise endurance, life quality, and improvement of symptoms. No serious adverse events were reported. So CS may be a promising treatment for patients with stable COPD of GOLD stages 2-3. No studies were placebo-controlled or of high methodological quality, which limits the conclusions.

18.
Article in English | MEDLINE | ID: mdl-30174706

ABSTRACT

Jianpiyifei II granule (JPYF II) is an oriental herbal formula used clinically in China to treat chronic obstructive pulmonary disease (COPD). The aim of the present study was to investigate the anti-inflammatory and antioxidative activities of JPYF II in a mouse model of COPD induced by lipopolysaccharide (LPS) and cigarette smoke (CS) and in RAW264.7 cells stimulated with cigarette smoke extract (CSE). Mice were given LPS via intratracheal instillation on days 1 and 15 and exposed to CS generated from 4 cigarettes/day for 28 days. The mice were treated with 0.75, 1.5, or 3 g/kg/d JPYF II by intragastric administration in low, middle, and high dose groups, respectively, for two weeks. RAW264.7 cells were stimulated by CSE and treated with JPYF II at doses of 12.5, 25, or 50 µg/mL. In the mouse model of LPS and CS-induced COPD, JPYF II decreased inflammatory cell counts in broncho alveolar lavage fluid (BALF), in addition to mRNA expression of proinflammatory cytokines and metalloproteinases (MMPs) in lung tissues. In addition, JPYF II elevated catalase (CAT) and glutathione peroxidase (GSH-Px) activities and reduced the levels of malondialdehyde (MDA) and IκBα and p65 phosphorylation and inflammatory cell infiltration in the lung tissues. In RAW264.7 cells stimulated with CSE, JPYF II inhibited the mRNA levels of inflammatory mediators and the phosphorylation of IκBα and p65. Our results suggest that JPYF II enhanced anti-inflammatory and antioxidative activities in a mouse model of COPD induced by LPS and CS and in RAW264.7 cells stimulated with CSE via inhibition of the NF-κB pathway.

19.
Sci Rep ; 7: 41288, 2017 01 23.
Article in English | MEDLINE | ID: mdl-28112272

ABSTRACT

Endothelial dysfunction is involved in the pathogenesis of decompression sickness (DCS) and contributes substantively to subsequent inflammatory responses. Escin, the main active compound in horse chestnut seed extract, is well known for its endothelial protection and anti-inflammatory properties. This study aimed to investigate the potential protection of escin against DCS in rats. Escin was administered orally to adult male rats for 7 d (1.8 mg/kg/day) before a simulated air dive. After decompression, signs of DCS were monitored, and blood and pulmonary tissue were sampled for the detection of endothelia related indices. The incidence and mortality of DCS were postponed and decreased significantly in rats treated with escin compared with those treated with saline (P < 0.05). Escin significantly ameliorated endothelial dysfunction (increased serum E-selectin and ICAM-1 and lung Wet/Dry ratio, decreased serum NO), and oxidative and inflammatory responses (increased serum MDA, MPO, IL-6 and TNF-α) (P < 0.05 or P < 0.01). The results suggest escin has beneficial effects on DCS related to its endothelia-protective properties and might be a drug candidate for DCS prevention and treatment.


Subject(s)
Decompression Sickness/drug therapy , Endothelial Cells/metabolism , Escin/therapeutic use , Protective Agents/therapeutic use , Animals , Antioxidants/pharmacology , Biomarkers/metabolism , Decompression Sickness/blood , Decompression Sickness/enzymology , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Escin/pharmacology , Inflammation/pathology , Male , Malondialdehyde/blood , Protective Agents/pharmacology , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism
20.
PLoS One ; 12(1): e0168881, 2017.
Article in English | MEDLINE | ID: mdl-28114372

ABSTRACT

Decompression sickness is a systemic pathophysiological process caused by bubbles and endothelial microparticles (EMPs) are established markers reflecting competency of endothelial function and vascular biology. Here, we investigated the effects of bubble-induced EMPs on endothelial cells in vitro and vivo. Rat pulmonary microvascular endothelial cells (PMVECs) were isolated and stimulated by bubbles and bubble-induced EMPs were collected and incubated with normal PMVECs in vitro. Cell viability and apoptosis were detected using Cell Counting Kit-8 assay and Annexin V FITC/PI double staining, respectively. Cell permeability and pro-inflammatory cytokines were determined by electric cell substrate impedance sensing and enzyme-linked immunosorbent assay, respectively. Intracellular nitric oxide and reactive oxygen species production were analyzed microscopically. In vivo study, bubble-induced EMPs were intravenously injected to the rats and soluble thrombomodulin, intercellular adhesion molecule 1, and vascullar adhesion molecule 1 were involved in evaluating endothelial dysfunction. In our study, bubble stimulus resulted in a significant increase of EMPs release by 3 fold. Bubble-induced EMPs significantly decreased cell viability and increased cell apoptosis. Moreover, bubble-induced EMPs induced abnormal increase of cell permeability and over-expression of pro-inflammatory cytokines. Intracellular ROS production increased while NO production decreased. These negative effects caused by bubble-induced EMPs were remarkably suppressed when EMPs pretreated with surfactant FSN-100. Finally, intravenous injection of bubble-induced EMPs caused elevations of soluble thrombomodulin and pro-inflammatory cytokines in the circulation. Altogether, our results demonstrated that bubble-induced EMPs can mediate endothelial dysfunction in vitro and vivo, which can be attenuated by EMPs abatement strategy. These data expanded our horizon of the detrimental effects of bubble-induced EMPs, which may be of great concern in DCS.


Subject(s)
Cell-Derived Microparticles , Endothelium, Vascular/physiopathology , Animals , Cells, Cultured , Male , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL