Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Sci Rep ; 13(1): 6833, 2023 04 26.
Article in English | MEDLINE | ID: mdl-37100790

ABSTRACT

To investigate the effect of pentoxifylline (PTX) on Chlorine (Cl2)-induced acute lung injury (ALI) by single-cell RNA sequencing (scRNA-seq). Female BALB/c mice were exposed to Cl2 at 400 ppm for 15 min. H&E staining was used to observe the degree of lung injury. scRNA-seq was conducted to analysis of normal and Cl2-exposed mice lung tissues. Immunofluorescence was used to observe genes of interest. Thirty-two mice were randomly divided into four groups: Control, Cl2, Cl2+Fer-1, Cl2+PTX. TEM, WB and ELISA were used to detect ferroptosis-related indicators. The 5, 8, 10, 12, 16, 20 clusters were epithelial cells and 4, 15, 18, 19, 21 clusters were endothelial cells. Pseudo-time analysis revealed the differentiation trajectory of epithelial cells and key regulatory genes (Gclc, Bpifa1, Dnah5 and Dnah9) during the process of injury. Cell-cell communication analysis identified several important receptor-ligand complexes (Nrp1-Vegfa, Nrp2-Vegfa, Flt1-Vegfa and Flt4-Vegfa). Ferroptosis were found up-regulated in epithelial and endothelial cells by GSVA analysis. Highly expressed genes to which closely related ferroptosis were found by SCENIC analysis. PTX could significantly decrease the levels of MDA and abnormal high expression of solute carrier family 7 member 11 (SLC7A11, the key transporter of cystine) as well as increase the expression of GSH/GSSG and glutathione peroxidase 4 (GPX4) (p < 0.05). This study revealed novel molecular features of Cl2-induced ALI. PTX may be a potential specific drug by inhibiting the process of ferroptosis in epithelial and endothelial cells.


Subject(s)
Acute Lung Injury , Ferroptosis , Pentoxifylline , Female , Animals , Mice , Chlorine/adverse effects , Pentoxifylline/adverse effects , Endothelial Cells , Transcriptome , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Acute Lung Injury/genetics , Glycoproteins , Phosphoproteins
2.
Environ Sci Pollut Res Int ; 30(19): 54742-54752, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36881233

ABSTRACT

This study researches the impact of terrain factors on chlorine gas diffusion processes based on SLAB model. Simulating the law of wind speed changing with altitude by calculating the real-time speed with vertical height combing actual terrain data, and integrating the influence of terrain on wind speed by using Reynolds Average Navier-Stokes (RANS) algorithm, K-turbulence model, and standard wall functions, then plotting the gas diffusion range in the map with terrain data according to the Gaussian-Cruger projection algorithm and dividing the hazardous areas according to the public exposure guidelines (PEG). The accidental chlorine gas releases near Lishan Mountain, Xi'an City, were simulated by the improved SLAB model. The results show that there are obvious differences analyzing contrastively the endpoint distance and area of chlorine gas dispersion under real terrain condition and ideal condition at different times; it can be found that the endpoint distance of the real terrain conditions is 1.34 km shorter than that of the ideal conditions at 300 s with terrain factors, and also the thermal area is 3,768,026m2 less than that of the ideal conditions. In addition, it can predict the specific number of casualties within different levels of harm at 2 min after chlorine gas dispersion, and casualties are constantly changing over time. The fusion of terrain factors can be used to optimize the SLAB model, which is expected to provide an important reference for effective rescue.


Subject(s)
Air Pollutants , Chlorine , Air Pollutants/analysis , Models, Theoretical , Computer Simulation , Wind
3.
BMC Pharmacol Toxicol ; 24(1): 12, 2023 02 27.
Article in English | MEDLINE | ID: mdl-36850013

ABSTRACT

OBJECTIVE: Chlorine is a chemical threat agent that can be harmful to humans. Inhalation of high levels of chlorine can lead to acute lung injury (ALI). Currently, there is no satisfactory treatment, and effective antidote is urgently needed. Pentoxifylline (PTX), a methylxanthine derivative and nonspecific phosphodiesterase inhibitor, is widely used for the treatment of vascular disorders. The present study was aimed to investigate the inhibitory effects of PTX on chlorine-induced ALI in rats. METHODS: Adult male Sprague-Dawley rats were exposed to 400 ppm Cl2 for 5 min. The histopathological examination was carried out and intracellular reactive oxygen species (ROS) levels were measured by the confocal laser scanning system. Subsequently, to evaluate the effect of PTX, a dose of 100 mg/kg was administered. The activities of superoxide dismutase (SOD) and the contents of malondialdehyde (MDA), glutathione (GSH), oxidized glutathione (GSSG) and lactate dehydrogenase (LDH) were determined by using commercial kits according to the manufacturer's instructions. Western blot assay was used to detect the protein expressions of SOD1, SOD2, catalase (CAT), hypoxia-inducible factor (HIF)-1α, vascular endothelial growth factor (VEGF), occludin, E-cadherin, bcl-xl, LC 3, Beclin 1, PTEN-induced putative kinase 1 (PINK 1) and Parkin. RESULTS: The histopathological examination demonstrated that chlorine could destroy the lung structure with hemorrhage, alveolar collapse, and inflammatory infiltration. ROS accumulation was significantly higher in the lungs of rats suffering from inhaling chlorine (P<0.05). PTX markedly reduced concentrations of MAD and GSSG, while increased GSH (P<0.05). The protein expression levels of SOD1 and CAT also decreased (P<0.05). Furthermore, the activity of LDH in rats treated with PTX was significantly decreased compared to those of non-treated group (P<0.05). Additionally, the results also showed that PTX exerted an inhibition effect on protein expressions of HIF-1α, VEGF and occludin, and increased the level of E-cadherin (P<0.05). While the up-regulation of Beclin 1, LC 3II/I, Bcl-xl, and Parkin both in the lung tissues and mitochondria, were found in PTX treated rats (P<0.05). The other protein levels were decreased when treated with PTX (P<0.05). CONCLUSION: PTX could ameliorate chlorine-induced lung injury via inhibition effects on oxidative stress, hypoxia and autophagy, thus suggesting that PTX could serve as a potential therapeutic approach for ALI.


Subject(s)
Acute Lung Injury , Pentoxifylline , Rats , Adult , Humans , Animals , Male , Rats, Sprague-Dawley , Chlorine , Pentoxifylline/pharmacology , Pentoxifylline/therapeutic use , Vascular Endothelial Growth Factor A , Glutathione Disulfide , Beclin-1 , Occludin , Reactive Oxygen Species , Superoxide Dismutase-1 , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Acute Lung Injury/prevention & control , Glutathione , Hypoxia , Ubiquitin-Protein Ligases
4.
Drug Chem Toxicol ; 46(6): 1100-1107, 2023 Nov.
Article in English | MEDLINE | ID: mdl-36220803

ABSTRACT

Inhalation of high concentrations of phosgene often causes pulmonary edema, which obstructs the airway and causes tissue hypoxia. There is currently no specific antidote. This study was performed to investigate the effect behind pentoxifylline (PTX) treatment for phosgene-induced lung injury in rat models. Rats were exposed to phosgene. The protein levels of hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor (VEGF), and occludin proteins in lung tissue were determined. The effect of both prophylactic and therapeutic administration of PTX (50 mg/kg and 100 mg/kg) was evaluated. The lung permeability index and HIF-1α protein level increased, the arterial blood oxygenation index (PaO2/FIO2 ratio) and occludin protein level decreased significantly 6 h after phosgene exposure (P < 0.05). PTX exerted protective effects by HIF-1α-VEGF-occludin signaling pathway to some extent. Moreover, prophylactic, but not therapeutic administration of PTX (100 mg/kg), exhibited a significant protective effect. Pretreatment with PTX protected against phosgene-induced lung injury, possibly by inhibiting differential expression of HIF-1α, VEGF, and occludin.


Subject(s)
Lung Diseases , Lung Injury , Pentoxifylline , Phosgene , Rats , Animals , Lung Injury/chemically induced , Lung Injury/drug therapy , Lung Injury/prevention & control , Pentoxifylline/pharmacology , Pentoxifylline/therapeutic use , Phosgene/toxicity , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Occludin/genetics , Vascular Endothelial Growth Factors , Hypoxia/chemically induced , Hypoxia/drug therapy
5.
Inhal Toxicol ; 34(13-14): 399-411, 2022.
Article in English | MEDLINE | ID: mdl-36260290

ABSTRACT

OBJECTIVE: Chlorine (Cl2), as an asphyxiant toxicant, induced poisoning incidents and acute lung injury (ALI) occur frequently. The specific pathogenesis of Cl2-induced ALI remains unclear. Immune cells play an important role in the process of lung damage. We used single-cell RNA sequencing (scRNA-seq) technology to explore T cells and macrophages molecular mechanism. METHODS: Female BALB/c mice were exposed to 400 ppm Cl2 for 15 min. scRNA-seq technology was used to observe the heterogeneity of T cells and macrophages. Hematoxylin-eosin (H&E) staining was used to evaluate the degree of lung injury. Immunofluorescence was used to verify the highly expressed genes of our interest. RESULTS: A total of 5316 to 7742 cells were classified into eight different cell types. Several new highly expressed anti-inflammatory and pro-inflammatory genes were found in T cells and macrophages, which were further verified in vitro. Through the pseudotime analysis of macrophages, it was found that the expression of pro-inflammatory and anti-inflammatory genes showed opposite trends in the development of Cl2-induced ALI. This study also mapped T cells-macrophage communication and identified the development of several important receptor-ligand complexes in Cl2-induced ALI. CONCLUSIONS: These findings are worthy of further exploration and provide new resources and directions for the study of Cl2-induced ALI in mice, especially in immune and inflammation mechanisms.


Subject(s)
Acute Lung Injury , Chlorine , Mice , Female , Animals , Chlorine/toxicity , T-Lymphocytes , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Lung/pathology , Mice, Inbred BALB C , Anti-Inflammatory Agents/pharmacology , Macrophages , Sequence Analysis, RNA , Lipopolysaccharides/toxicity
6.
Biomed Pharmacother ; 108: 1181-1187, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30372819

ABSTRACT

Obesity, diabetes and related metabolic disorders are among the top prevalent metabolism-related diseases with increasing threat to human health throughout the world. Oleanolic acid (OA) is a natural triterpenoid and an aglycone of many saponins possessing anti-diabetic, antioxidant, hypolipidemic and anti-inflammatory activities. A nano-formulation of OA was recently developed to evaluate the efficiency of nano-OA in the treatment of insulin-resistance and metabolic disorders in high fat and fructose (HFF) diet-fed rats. This study further identified that nano-OA could reduce the increase of body weights, serum insulin, insulin sensitivity index, serum triglycerides, and cholesterol in HFF-fed rats. In consistence, nano-OA was able to attenuate HFF diet-induced lipid accumulation in the liver and improve the structural integrity of mitochondria and endoplasmic reticulum in liver and pancreas in animals fed with HFF diet. In addition, nan-OA can efficaciously mitigate the increase of levels of malondialdehyde (MDA) and nitric oxide (NO), and serum superoxide dismutase (SOD) and catalase (CAT) activities in blood samples. The beneficial effects of nano-OA was further evidenced to be superior to OA formulated in arabic gum and rosiglitazone treatment. Together, this study provides the evidence that nano-OA can effectively improve HFF diet-induced metabolic dysfunctions in rats by improving its bioavailability and pharmacodynamic properties and thus nano-OA may be a potentially efficient agent to treat obesity-related diabetes and metabolic disorders.


Subject(s)
Metabolic Diseases/drug therapy , Nanoparticles/chemistry , Oleanolic Acid/therapeutic use , Animals , Diet, High-Fat , Fructose , Insulin Resistance , Lipid Metabolism/drug effects , Liver/injuries , Liver/metabolism , Liver/pathology , Liver/ultrastructure , Male , Metabolic Diseases/pathology , Oleanolic Acid/pharmacology , Oxidative Stress/drug effects , Pancreas/drug effects , Pancreas/injuries , Pancreas/pathology , Pancreas/ultrastructure , Rats, Sprague-Dawley
7.
Mol Cancer Ther ; 14(9): 2090-102, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26141949

ABSTRACT

Tumor adaptive resistance to therapeutic radiation remains a barrier for further improvement of local cancer control. SIRT3, a member of the sirtuin family of NAD(+)-dependent protein deacetylases in mitochondria, promotes metabolic homeostasis through regulation of mitochondrial protein deacetylation and plays a key role in prevention of cell aging. Here, we demonstrate that SIRT3 expression is induced in an array of radiation-treated human tumor cells and their corresponding xenograft tumors, including colon cancer HCT-116, glioblastoma U87, and breast cancer MDA-MB231 cells. SIRT3 transcriptional activation is due to SIRT3 promoter activation controlled by the stress transcription factor NF-κB. Posttranscriptionally, SIRT3 enzymatic activity is further enhanced via Thr150/Ser159 phosphorylation by cyclin B1-CDK1, which is also induced by radiation and relocated to mitochondria together with SIRT3. Cells expressing Thr150Ala/Ser159Ala-mutant SIRT3 show a reduction in mitochondrial protein lysine deacetylation, Δψm, MnSOD activity, and mitochondrial ATP generation. The clonogenicity of Thr150Ala/Ser159Ala-mutant transfectants is lower and significantly decreased under radiation. Tumors harboring Thr150Ala/Ser159Ala-mutant SIRT3 show inhibited growth and increased sensitivity to in vivo local irradiation. These results demonstrate that enhanced SIRT3 transcription and posttranslational modifications in mitochondria contribute to adaptive radioresistance in tumor cells. CDK1-mediated SIRT3 phosphorylation is a potential effective target to sensitize tumor cells to radiotherapy.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Mitochondria/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Radiation Tolerance/genetics , Sirtuin 3/genetics , Transcriptional Activation , Acetylation , Animals , CDC2 Protein Kinase , Cell Line, Tumor , Disease Models, Animal , Enzyme Activation , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Mitochondria/radiation effects , Mitochondrial Proteins/metabolism , Mutation , NF-kappa B/metabolism , Neoplasms/pathology , Neoplasms/radiotherapy , Phosphorylation , Sirtuin 3/metabolism , Transcription, Genetic
8.
Antioxid Redox Signal ; 22(11): 921-37, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25621814

ABSTRACT

AIMS: The liver undergoes marked changes in the rate of proliferation during normal development and regeneration through the coordinated activity of numerous signaling pathways. Little is known, however, about the events that act upstream of these signaling pathways. Here, we explore the modulatory effects of hydrogen peroxide (H2O2) on these pathways in the context of liver development and regeneration. RESULTS: We show that H2O2 production during liver development and after partial hepatectomy is tightly regulated in time by specific H2O2-producing and scavenging proteins and dose dependently triggers two distinct pathways. Sustained elevated H2O2 levels are required for the activation of ERK signaling and trigger a shift from quiescence to proliferation. Contrastingly, sustained decreased H2O2 levels are required for the activation of p38 signaling and trigger a shift from proliferation to quiescence. Both events impact the cyclin D and Rb pathways and are involved in liver development and regeneration. Pharmacological lowering of H2O2 levels reduces the extent of fetal hepatocyte proliferation and delays the onset of liver regeneration. Chemical augmentation of H2O2 levels in adult hepatocytes triggers proliferation and delays the termination of liver regeneration. INNOVATION: Our results challenge the traditional view of H2O2 as a deleterious stressor in response to liver damage and identify a novel role of endogenous H2O2 in liver development and regeneration. CONCLUSIONS: Endogenous H2O2 production is tightly regulated during liver development and regeneration. H2O2 constitutes an important trigger for the proliferation and quiescence transition in hepatocytes via the concentration-dependent activation of the ERK or p38 pathway.


Subject(s)
Cell Proliferation/drug effects , Hydrogen Peroxide/pharmacology , Liver Regeneration/drug effects , Liver/drug effects , Oxidants/pharmacology , Animals , Animals, Newborn , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Glucose/metabolism , Hepatectomy , Hepatocytes/metabolism , Liver/embryology , Liver/growth & development , Male , Rats, Sprague-Dawley , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Oxid Med Cell Longev ; 2014: 320513, 2014.
Article in English | MEDLINE | ID: mdl-24669284

ABSTRACT

Reactive oxygen species (ROS) are closely related to the aging process. In our previous studies, we found that the saponins from Aralia taibaiensis have potent antioxidant activity, suggesting the potential protective activity on the aging. However, the protective effect of the saponins and the possible underlying molecular mechanism remain unknown. In the present study, we employed a D-galactose-induced aging rat model to investigate the protective effect of the saponins. We found that D-galactose treatment induced obvious aging-related changes such as the decreased thymus and spleen coefficients, the increased advanced glycation end products (AGEs) level, senescence-associated ß-galactosidase (SAß-gal) activity, and malondialdehyde (MDA) level. Further results showed that Forkhead box O3a (FOXO3a), nuclear factor-erythroid 2-related factor 2 (Nrf2), and their targeted antioxidants such as superoxide dismutase 2 (SOD2), catalase (CAT), glutathione reductase (GR), glutathione (GSH), glutamate-cysteine ligase (GCL), and heme oxygenase 1 (HO-1) were all inhibited in the aging rats induced by D-galactose treatment. Saponins supplementation showed effective protection on these changes. These results demonstrate that saponins from Aralia taibaiensis attenuate the D-galactose-induced rat aging. By activating FOXO3a and Nrf2 pathways, saponins increase their downstream multiple antioxidants expression and function, at least in part contributing to the protection on the D-galactose-induced aging in rats.


Subject(s)
Aging/drug effects , Aralia/chemistry , Forkhead Transcription Factors/metabolism , NF-E2-Related Factor 2/metabolism , Saponins/pharmacology , Signal Transduction/drug effects , Animals , Antioxidants/metabolism , Forkhead Box Protein O3 , Galactose , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Malondialdehyde/metabolism , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Spleen/drug effects , Spleen/pathology , Thymus Gland/drug effects , Thymus Gland/pathology
10.
Toxicol Lett ; 224(2): 165-74, 2014 Jan 13.
Article in English | MEDLINE | ID: mdl-24188932

ABSTRACT

Arsenic exposure has been shown to induce hypoxia inducible factor 1α (HIF-1α) accumulation, however the underlying mechanism remains unknown. In the present study, we tested the hypothesis that arsenic exposure triggered the interaction between NADPH oxidase and mitochondria to promote reactive oxygen species (ROS) production, which inactivate prolyl hydroxylases (PHDs) activity, leading to the stabilization of HIF-1α protein. Exposure of human immortalized liver cell line HL-7702 cells to arsenite induced HIF-1α accumulation in a dose-dependent manner, which was abolished by SOD mimetic MnTMPyP. Inhibition of NADPH oxidase with diphenyleneiodonium chloride (DPI) or inhibition of mitochondrial respiratory chain with rotenone significantly blocked arsenite-induced ROS production, and the mitochondria appeared to be the major source of ROS production. Arsenite treatment inhibited HIF-1α hydroxylation by prolyl hydroxylases (PHDs) and increased HIF-1α stabilization, but did not affect HIF-1α mRNA expression and Akt activation. Supplementation of ascorbate or Fe(II) completely abolished arsenite-induced PHDs inhibition and HIF-1α stabilization. In conclusion, these results define a unique mechanism of HIF-1α accumulation following arsenic exposure, that is, arsenic activates NADPH oxidase-mitochondria axis to produce ROS, which deplete intracellular ascorbate and Fe(II) to inactivate PHDs, leading to HIF-1α stabilization.


Subject(s)
Arsenites/toxicity , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mitochondria/physiology , NADPH Oxidases/physiology , Prolyl-Hydroxylase Inhibitors/toxicity , Reactive Oxygen Species/metabolism , Cells, Cultured , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/chemistry
11.
Chem Biol Interact ; 207: 7-15, 2014 Jan 25.
Article in English | MEDLINE | ID: mdl-24211618

ABSTRACT

Combinations of antioxidants are believed to be more effective than single antioxidant because when antioxidants are combined they support each other synergistically to create a magnified effect. Discovering the enhancer effects or synergies between bioactive components is valuable for resisting oxidative stress and improving health benefits. The aim of this study was to investigate a possible cooperation of natural antioxidant caffeic acid phenethyl ester (CAPE) with synthetic antioxidant Trolox in the model systems of chemical generation of free radicals, lipid peroxidation of microsomes and radiation-induced oxidative injury in L929 cells. Based on the intermolecular interaction between CAPE and Trolox, the present study shows a synergistic effect of CAPE and Trolox in combination on elimination of three different free radicals and inhibition of lipid peroxidation initiated by three different systems. CAPE and Trolox added simultaneously to the L929 cells exerted an enhanced preventive effect on the oxidative injury induced by radiation through decreasing ROS generation, protecting plasma membrane and increasing the ratios of reduced glutathione/oxidized glutathione and the expression of key antioxidant enzymes mediated by nuclear factor erythroid 2 p45-related factor 2 (Nrf2). Our results showed for the first time that administration of CAPE and Trolox in combination may exert synergistic antioxidant effects, and further indicate that CAPE and Trolox combination functions mainly through scavenging ROS directly, inhibiting lipid peroxidation and promoting redox cycle of GSH mediated by Nrf2-regulated glutathione peroxidase and glutathione reductase expression.


Subject(s)
Antioxidants/pharmacology , Caffeic Acids/pharmacology , Chromans/pharmacology , Gamma Rays , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Phenylethyl Alcohol/analogs & derivatives , Animals , Biphenyl Compounds/metabolism , Cell Line , Cell Survival/drug effects , Cell Survival/radiation effects , Free Radical Scavengers/pharmacology , Glutathione Disulfide/metabolism , Lipid Peroxidation/drug effects , Lipid Peroxidation/radiation effects , Male , Mice , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Microsomes, Liver/radiation effects , Phenylethyl Alcohol/pharmacology , Picrates/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Spectrophotometry, Ultraviolet , Thiobarbituric Acid Reactive Substances/metabolism
12.
Exp Toxicol Pathol ; 65(3): 311-8, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22030112

ABSTRACT

The mechanism of phosgene-induced acute lung injury (ALI) remains unclear and it is still lack of effective treatments. Previous study indicated that oxidative stress was involved in phosgene-induced ALI. Caffeic acid phenethyl ester (CAPE) has been proved to be an anti-inflammatory agent and a potent free radical scavenger. The purpose of this study was to investigate the protective effects of CAPE on phosgene-induced ALI and identify the mechanism, in which oxidative stress and inflammation were involved. The phosgene was used to induce ALI in rats. The results showed that after phosgene exposure, total protein content in BALF was not significantly changed. The increase of MDA level and SOD activity induced by phosgene was significantly reduced by CAPE administration, and the decrease of GSH level in BALF and lung were significantly reversed by CAPE. CAPE also partially blocked the translocation of NF-κB p65 to the nucleus, but it had little effect on the phosphorylation of p38 MAPK. In conclusion, CAPE showed protective effects on lung against phosgene-induced ALI, which may be related with a combination of the antioxidant and anti-inflammatory functions of CAPE.


Subject(s)
Acute Lung Injury/prevention & control , Air Pollutants/toxicity , Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Caffeic Acids/therapeutic use , Phosgene/toxicity , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Animals , Anti-Inflammatory Agents/administration & dosage , Antioxidants/administration & dosage , Blotting, Western , Bronchoalveolar Lavage Fluid/chemistry , Caffeic Acids/administration & dosage , Lung/drug effects , Lung/enzymology , Lung/immunology , Male , Malondialdehyde/metabolism , Organ Size/drug effects , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism , Transcription Factor RelA/metabolism
13.
J Appl Toxicol ; 33(1): 71-7, 2013 Jan.
Article in English | MEDLINE | ID: mdl-21818760

ABSTRACT

Phosgene is a poorly water-soluble gas penetrating the lower respiratory tract which can induce acute lung injury characterized by a latent phase of fatal pulmonary edema. Pulmonary edema caused by phosgene is believed to be a consequence of oxidative stress and inflammatory responses. Ethyl pyruvate (EP) has been demonstrated to have anti-inflammatory and anti-oxidative properties in vivo and in vitro. The potential therapeutic role of EP in phosgene-induced pulmonary edema has not been addressed so far. In the present study, we aim to investigate the protective effects of EP on phosgene-induced pulmonary edema and the underlying mechanisms. Rats were administered with EP (40 mg kg(-1)) and RAW264.7 cells were also incubated with it (0, 2, 5 or 10 µm) immediately after phosgene (400 ppm, 1 min) or air exposure. Wet-to-dry lung weight ratio (W:D ratio), nitric oxide (NO) and prostaglandin E(2) (PGE(2)) production, cyclooxygenase2 (COX-2) and inducible nitric oxide synthase (iNOS) expression, and mitogen-activated protein kinases activities (MAPKs) were measured. Our results showed that EP treatment attenuated phosgene-induced pulmonary edema and decreased the level of NO and PGE(2) dose-dependently. Furthermore, EP significantly reduced COX-2 expression, iNOS expression and MAPK activation induced by phosgene. Moreover, specific inhibitors of MAPKs reduced COX-2 and iNOS expression induced by phosgene. These findings suggested that EP has a protective role against phosgene-induced pulmonary edema, which is mediated in part by inhibiting MAPK activation and subsequently down-regulating COX-2 and iNOS expression as well as decreasing the production of NO and PGE(2).


Subject(s)
Lung/drug effects , Phosgene/toxicity , Protective Agents/pharmacology , Pulmonary Edema/prevention & control , Pyruvates/pharmacology , Animals , Bronchoalveolar Lavage Fluid/chemistry , Cell Line , Chemical Warfare Agents/toxicity , Cyclooxygenase 2/metabolism , Lung/pathology , Macrophages/drug effects , Macrophages/pathology , Male , Mitogen-Activated Protein Kinases/metabolism , Nitric Oxide/analysis , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Organ Size/drug effects , Pulmonary Edema/chemically induced , Pulmonary Edema/pathology , Rats , Rats, Sprague-Dawley
14.
Toxicol Sci ; 127(1): 120-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22387748

ABSTRACT

Human arsenic exposure is associated with increased risk of skin cancer, and arsenite greatly enhances ultraviolet (UV)-induced skin tumors in a mouse model of carcinogenesis. Inhibition of DNA repair is one proposed mechanism for the observed cocarcinogenicity. We have previously demonstrated that low concentrations of arsenite inhibit poly(ADP-ribose) polymerase (PARP)-1, thus interfering with DNA repair process triggered by UV radiation. Because overactivation of PARP-1 often leads to apoptotic cell death, and unrepaired DNA lesions promote genomic instability and carcinogenesis, we hypothesized that inhibition of PARP-1 by arsenic may promote the survival of potentially "initiated carcinogenic cells," i.e., cells with unrepaired DNA lesions. In the present study, we tested this hypothesis on UV-challenged HaCat cells. Cells were pretreated with 2µM arsenite for 24 h before UV exposure. Outcome parameters included apoptotic death rate, PARP-1 activation, apoptotic molecules, and retention of DNA lesions. UV exposure induced PARP-1 activation and associated poly(ADP-ribose) production, apoptosis-inducing factor release, cytochrome C release, and caspases activation, which led to apoptotic death in HaCat cells. Pretreatment with 2µM arsenite significantly inhibited UV-induced cell death as well as the associated molecular events. Notably, knockdown of PARP-1 with small interfering RNA completely abolished the antagonism of arsenite. Furthermore, arsenite pretreatment led to long-term retention of UV-induced cyclobutane pyrimidine dimers. Together, these results suggest that low concentration of arsenite reduces UV-induced apoptosis via inhibiting PARP-1, thus promoting the survival of cells with unrepaired DNA lesions, which may be an important mechanism underlying arsenic cocarcinogenic action.


Subject(s)
Arsenites/toxicity , DNA Damage/drug effects , DNA Repair/drug effects , Enzyme Inhibitors/toxicity , Poly(ADP-ribose) Polymerase Inhibitors , Ultraviolet Rays/adverse effects , Apoptosis/drug effects , Apoptosis/radiation effects , Caspase Inhibitors , Cell Line, Transformed , Cell Survival/drug effects , Dose-Response Relationship, Drug , Gene Knockdown Techniques , Gene Silencing , Humans , Keratinocytes/drug effects , Keratinocytes/radiation effects , Poly (ADP-Ribose) Polymerase-1 , Radiation Effects
15.
Carbohydr Polym ; 89(3): 890-8, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-24750877

ABSTRACT

Oxidative stress is associated with insulin resistance (IR) and is thought to contribute to the development and progression toward type 2 diabetes (T2DM). This study was undertaken to isolate the bioactive polysaccharide (SMPW1) from Salvia miltiorrhiza Bunge and investigated its protective effects on IR model in rats induced by tert-butyl hydroperoxide (t-BHP). In vivo animal experiments showed that SMPW1 (50 and 100mg/kg) possessed high antioxidative and protective capacity against the injury induced by t-BHP, as reflected in the increased expression or activities of catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GPx), and the decreased formation of malondialdehyde (MDA) in serum and liver homogenates. In addition, SMPW1 (50 and 100mg/kg) also attenuated IR and the morphological injury of liver and pancreas induced by t-BHP, and improved insulin sensitivity index. In conclusion, SMPW1 can protect against the development of T2DM and improve IR via reduction of oxidative stress.


Subject(s)
Insulin Resistance , Polysaccharides/pharmacology , Salvia miltiorrhiza/chemistry , Animals , Male , Rats , Rats, Sprague-Dawley
16.
Inhal Toxicol ; 23(13): 842-52, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22035124

ABSTRACT

The principal acute mode of action of inhaled phosgene gas is related to an increase alveolar fluid exudation under pathologic conditions. This paper considers some aspects in modeling phosgene-induced acute lung injury (ALI) in an acute rat bioassay and whether edema formation can be modulated by inhaled nitric oxide (iNO). Protein analysis in bronchoalveolar lavage (BAL) fluid is amongst the most sensitive method to quantify the phosgene-induced non-cardiogenic, pulmonary high-permeability edema following acute inhalation exposure. Maximum concentrations in BAL-protein occur within one day postexposure, typically within a latency period up to about 15 h as a consequence of an increasingly exhausted lymphatic drainage. An almost similar sensitivity was given by the functional endpoint 'enhanced pause (Penh)' when measured by non-invasive whole-body barometric plethysmography over a time period of 20 h. The magnitude of edema formation follows a concentration x time (C¹xt) relationship, although animal model-specific deviations may occur at very short exposure durations (1-20 min) due to a rodent-specific, reflexively induced transient decreased ventilation. This has to be accounted for when simulating accidental exposure scenarios to study the mechanisms involved in pharmacological modulation of fluid transport in this type of ALI. Therefore, a special focus has to be given to the dosimetry of inhaled phosgene, otherwise any change in effect magnitude, as a result of under-dosing of phosgene, may be misconceived as promising therapy. This study demonstrates that accidental exposures can be modeled best in rats by exposure durations of at least 20-30 min. Lung function measurements (Penh) show that pathophysiological effects appear to occur concomitant with the exposure to phosgene; however, its full clinical manifestation requires a gross imbalance of pulmonary fluid clearance. When applying this concept, post-phosgene exposure iNO at 1.5 ppm × 6 h or 15 pm × 20 h led to an aggravation of edema formation while L-NAME, a non-selective inhibitor of nitric oxide synthase, led to attenuation. Ethyl pyruvate, given either prophylactically or therapeutically, was ineffective.


Subject(s)
Acute Lung Injury/chemically induced , Irritants , Nitric Oxide/toxicity , Phosgene , Acute Lung Injury/drug therapy , Acute Lung Injury/physiopathology , Animals , Bronchoalveolar Lavage Fluid/chemistry , Disease Models, Animal , Drug Synergism , Enzyme Inhibitors/therapeutic use , Male , NG-Nitroarginine Methyl Ester/therapeutic use , Nitric Oxide Synthase/antagonists & inhibitors , Plethysmography, Whole Body , Proteins/analysis , Pyruvates/therapeutic use , Rats , Rats, Wistar
17.
Food Chem Toxicol ; 49(11): 2750-7, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21843584

ABSTRACT

Myocardial ischemia/reperfusion (MI/R) is a major cause for the events of cardiovascular disease. Oxidative stress plays a critical role in the development of ischemia/reperfusion (IR) injury. As a potent antioxidant, alpha-lipoic acid (LA) has been shown to provide a benefit for the inhibition of IR injury and inhibit reactive oxygen species (ROS) generation during MI/R in rats. However, the mechanism on the protective effect of LA is still to be clarified. The present study was aimed to investigate the protective effect of LA against MI/R injury and its mechanisms. We found that 2h of myocardial ischemia followed by different time periods of reperfusion resulted in significant increase of creatine kinase (CK) activity. MI/R also significantly promoted oxidative stress and decreased the activities of antioxidant enzymes. In addition, apoptosis and inflammatory response were activated and aggravated in a time-dependent manner by MI/R. All these alterations induced by MI/R were attenuated by the administration of LA 30 min before reperfusion. These results suggested that LA played a protective effect against MI/R injury via antioxidant, anti-apoptotic and anti-inflammatory effects. These findings may significantly better the understanding of the pharmacological actions of LA and advance therapeutic approaches to MI/R injury and cardiovascular diseases.


Subject(s)
Antioxidants/pharmacology , Myocardial Ischemia/prevention & control , Reperfusion Injury/prevention & control , Thioctic Acid/pharmacology , Animals , Apoptosis , Blotting, Western , Catalase/metabolism , Creatine Kinase/metabolism , Glutathione , Male , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
18.
Phytother Res ; 25(7): 1031-40, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21254272

ABSTRACT

Diabetes mellitus (DM) is a metabolic disorder characterized by chronic hyperglycemia. Although the clear mechanisms of DM and insulin resistance are still to be cleared, it has been well documented that reactive oxygen species (ROS) play a pivotal role in DM and multiple types of insulin resistance. For the past few years, natural substances have been shown to have the potential to treatment DM. Attention has been especially focused on plants rich in triterpenoids, which generally show antioxidant and antiglycation effect. In our previous studies, it was shown that oleanolic acid (OA), a natural triterpenoid and an aglycone of many saponins, is a potent antioxidant acting as not only a free radical-scavenger through direct chemical reactions but also as a biological molecule, which may enhance the antioxidant defenses. The present study aimed to investigate the potential antidiabetic effect of OA. Oleanolic acid showed a significant blood glucose-lowering and weight-losing effect in diabetic animals induced by streptozotocin (STZ). In the insulin resistant model, it was also shown that OA may promote insulin signal transduction and inhibit oxidative stress-induced hepatic insulin resistance and gluconeogenesis, in which process the phosphorylation of ERK and the protective effect on mitochondrial function may be involved. These findings may significantly better the understanding of the pharmacological actions of OA and advance therapeutic approaches to DM.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Hypoglycemic Agents/pharmacology , Oleanolic Acid/pharmacology , Animals , Antioxidants/pharmacology , Blood Glucose/drug effects , Cell Line , Hepatocytes/drug effects , Humans , Insulin/metabolism , Insulin Resistance , Male , Membrane Potential, Mitochondrial , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/pharmacology , Signal Transduction/drug effects
19.
Exp Toxicol Pathol ; 63(6): 527-33, 2011 Sep.
Article in English | MEDLINE | ID: mdl-20510595

ABSTRACT

Tert-butyl hydroperoxide (t-BHP) can induce cell injury by forming free radical intermediates. Peroxisome proliferator-activated receptor (PPAR)-γ is a ligand-activated transcription factor belonging to nuclear hormone receptor superfamily, and is involved in oxidative stress response. Thiazolidinedione rosiglitazone is a potent PPARγ agonist. The main aim of this study was to investigate the protective effect of rosiglitazone on QZG cells from t-BHP-induced toxicity. MTT assay showed that t-BHP treatment resulted in decreased cell viability in a concentration dependent manner. Under 400 µM t-BHP treatment, QZG cell displayed significant loss of viability and dramatic morphological changes characterized by changing in shape from triangle to spherical, disappearance of cell cilia, swollen mitochondrial and typical apoptotic alteration such as condensation of chromatin, and appearance of crescent under light microscopy and electronic microscopy, respectively. Flow cytometry analysis indicated that 30.90±1.70% QZG cells were undergoing apoptosis compared to that of the control cells (2.80±0.85%, P<0.05). There was substantial population of the cells undergoing necrosis (28.5.%). 25 µM rosiglitazone treatment inhibited the t-BHP-induced cell toxicity significantly by restoring the cell viability, reducing cell population undergone apoptosis to normal level (3.5%) and ameliorating t-BHP-induced pathological changes. Real-time RT-PCR results showed that 400 µM t-BHP caused dramatic down-regulation of PPARγ expression in QZG cells, whereas combining treatment with 25 µM rosiglitazone resistant to PPARγ expression to normal level partially. Overall, our results indicate that rosiglitazone has protective effect against t-BHP-induced QZG cell injury. The protective effect of rosiglitazone is involved in its regulation on the function of PPARγ.


Subject(s)
Antioxidants/pharmacology , Oxidative Stress/drug effects , PPAR gamma/agonists , Thiazolidinediones/pharmacology , tert-Butylhydroperoxide/toxicity , Cell Culture Techniques , Cell Line , Cell Survival/drug effects , Data Interpretation, Statistical , Flow Cytometry , Humans , Microscopy, Electron, Transmission , Microscopy, Phase-Contrast , PPAR gamma/biosynthesis , Rosiglitazone
20.
Inhal Toxicol ; 22(11): 889-95, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20569121

ABSTRACT

Phosgene inhalation results in acute lung injury (ALI) mostly, pulmonary edema and even acute respiratory distress syndrome, but there is no specific antidote. Inflammatory cells play an important role in the ALI caused by phosgene. Intercellular adhesion molecule-1 (ICAM-1) is a critical factor for inflammatory organ injury. We hypothesized that pentoxifylline (PTX), an inhibitor of leukocyte activation, would have a protective effect on experimental phosgene-induced lung injury rats by inhibiting ICAM-1. To prove this hypothesis, we used rat models of phosgene (400 ppm x 1 min)-induced injury to investigate: (1) the time course of lung injury (control 1, 3, 6, 12, 24, and 48 h group), including pathological changes in hematoxylin and eosin staining and transmission electron microscope, myeloperoxidase (MPO) activity by colorimetric method and ICAM-1 protein level detected by western blot, (2) At 3 h after phosgene exposure, protective effects of different dosages of PTX (50 mg/kg and 100 mg/kg) administration were evaluated by MPO activity, ICAM-1 differential expression and WBC count in bronchoalveolar lavage fluid. The results showed that inflammatory cells emerged out of lung blood vessels at 3 h after phosgene exposure. The MPO activity of lung tissue increased significantly from 3 to 48 h after phosgene exposure (P < 0.05) and ICAM-1 expression presented a similar change, especially at 3 h and 24 h (P < 0.05). After pretreatment and treatment with PTX (100 mg/kg), significant protective effects were shown (P < 0.05). These data supported our hypothesis that PTX reduced phosgene-induced lung injury, possibly by inhibiting ICAM-1 differential expression.


Subject(s)
Intercellular Adhesion Molecule-1/biosynthesis , Lung Injury/metabolism , Lung Injury/prevention & control , Pentoxifylline/pharmacology , Pentoxifylline/therapeutic use , Phosgene/toxicity , Animals , Inhalation Exposure/adverse effects , Lung Injury/chemically induced , Male , Peroxidase/antagonists & inhibitors , Peroxidase/metabolism , Phosgene/administration & dosage , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...