Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 118
Filter
Add more filters











Publication year range
1.
Comput Math Methods Med ; 2020: 6374014, 2020.
Article in English | MEDLINE | ID: mdl-33123215

ABSTRACT

OBJECTIVE: In recent years, pulmonary fibrosis caused by paraquat poisoning is still concerned. However, no effective drugs have been developed yet to treat paraquat-induced pulmonary fibrosis. The aim of our research is to investigate whether imrecoxib can inhibit paraquat-induced pulmonary fibrosis and its possible mechanism. METHODS: Extraction of primary pulmonary fibrosis cells (PPF cells) in vitro by the method of trypsin digestion. RT-qPCR and western blot were employed to measure the transcription level and protein expression of EMT related markers in paraquat-induced A549 cells. MTT, wound-healing, and Transwell experiments were used to verify the effect of imrecoxib on the proliferation, migration, and invasion of PPF and HFL1 cells. RESULTS: Firstly, our results confirmed that paraquat can induce EMT and activate the NF-κB/snail signal pathway in lung epithelial cell A549. Furthermore, experimental results showed that imrecoxib could repress the proliferation, migration, and invasion of PPF and HFL1 cells. Finally, our study found that imrecoxib can inhibit EMT of paraquat-induced A549 cells by the NF-κB/snail signal pathway. CONCLUSION: Imrecoxib can inhibit EMT of paraquat-induced A549 cells and alleviate paraquat-caused pulmonary fibrosis through the NF-κB/snail signal pathway. Therefore, imrecoxib is a drug worthy of study in the treatment of paraquat-induced pulmonary fibrosis.


Subject(s)
Paraquat/antagonists & inhibitors , Paraquat/poisoning , Pulmonary Fibrosis/drug therapy , Pyrroles/pharmacology , Sulfides/pharmacology , A549 Cells , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Computational Biology , Cyclooxygenase 2 Inhibitors/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Herbicides/antagonists & inhibitors , Herbicides/poisoning , Humans , Lung/drug effects , Lung/metabolism , Lung/pathology , Mathematical Concepts , NF-kappa B/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Signal Transduction/drug effects , Snail Family Transcription Factors/metabolism
2.
Toxicology ; 426: 152267, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31381934

ABSTRACT

Paraquat has relatively strong detrimental effects on humans and animals and can cause acute lung injury with high mortality. Ghrelin is a brain-gut peptide which plays important roles in regulating various physiological processes. This study investigated whether ghrelin could inhibit paraquat-induced lung injuries and attempted to elucidate the possible molecular mechanisms. A549 cells were preincubated with different concentrations of ghrelin and then treated with 200 µM of PQ for 24 h. Then cell survival, apoptosis, cellular oxidative stress and lipid peroxidation of A549 cells were detected after different treatments. Subsequently, we analyzed the mitochondrial membrane potential (ΔΨm) and measured caspase-3 activation in A549 cells. In addition, we investigated the activation of the MAPKs pathway and the function of p38-MAPK within mitochondrial apoptosis. Our study indicated that ghrelin administration improved cell viability and reduced apoptosis of PQ-treated A549 cells dose-dependently. Ghrelin treatment reduced the elevation of ROS and MDA, while improved GSH content in A549 cells after paraquat exposure. Moreover, we found that ghrelin dose-dependently increased ΔΨm and decreased caspase-3 activity. The phosphorylated p38 MAPK and JNK levels elevated following PQ exposure, while the phosphorylation of p38 MAPK decreased following ghrelin pretreatment. p38 MAPK siRNA or SB203580 pretreatment ameliorated PQ-caused cell injury and apoptosis related signals, however, the intracellular ROS production was not affected. N-Acetylcysteine (NAC), a classic antioxidant pretreatment decreased the phosphorylated p38 MAPK level and intracellular ROS production, alleviated cell injury, and inhibited apoptosis. The results showed that p38-MAPK pathway plays an important role in PQ-caused alveolar epithelial cell insult, and ghrelin might attenuate PQ-induced cell injury by inhibiting ROS-induced p38-MAPK modulated mitochondrial apoptotic pathway.


Subject(s)
Apoptosis/drug effects , Ghrelin/pharmacology , Herbicides/toxicity , MAP Kinase Signaling System/drug effects , Mitochondria/drug effects , Paraquat/antagonists & inhibitors , Paraquat/toxicity , p38 Mitogen-Activated Protein Kinases/metabolism , A549 Cells , Caspase 3/biosynthesis , Caspase 3/drug effects , Cell Survival/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/drug effects
3.
Aging (Albany NY) ; 11(15): 5726-5743, 2019 08 12.
Article in English | MEDLINE | ID: mdl-31403933

ABSTRACT

Carbohydrate overconsumption increases blood glucose levels, which contributes to the development of various diseases including obesity and diabetes. It is generally believed that high glucose metabolism increases cellular reactive oxygen species (ROS) levels, damages insulin-secreting cells and leads to age-associated diabetic phenotypes. Here we find that in contrast, high glucose suppresses ROS production induced by paraquat in both mammalian cells and the round worm C. elegans. The role of glucose in suppressing ROS is further supported by glucose's ability to alleviate paraquat's toxicity on C. elegans development. Consistently, we find that the ROS-regulated transcription factor SKN-1 is inactivated by glucose. As a result, the ROS/SKN-1-dependent lifespan extension observed in paraquat-treated animals, mitochondrial respiration mutant isp-1 and germline-less mutant glp-1 are all suppressed by glucose. Our study reveals an unprecedented interaction of glucose with ROS, which could have significant impact on our current understanding of glucose- and ROS-related diseases.


Subject(s)
Caenorhabditis elegans/metabolism , Glucose/metabolism , Longevity/physiology , Reactive Oxygen Species/metabolism , Aging/drug effects , Aging/genetics , Aging/metabolism , Animals , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cell Death/drug effects , Cell Death/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Glucose/pharmacology , Herbicides/antagonists & inhibitors , Herbicides/toxicity , Humans , Longevity/drug effects , Models, Biological , Oxidative Stress/drug effects , Paraquat/antagonists & inhibitors , Paraquat/toxicity , Species Specificity , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Aging Cell ; 18(5): e12990, 2019 10.
Article in English | MEDLINE | ID: mdl-31264342

ABSTRACT

Paraquat (PQ) promotes cell senescence in brain tissue, which contributes to Parkinson's disease. Furthermore, PQ induces heart failure and oxidative damage, but it remains unknown whether and how PQ induces cardiac aging. Here, we demonstrate that PQ induces phenotypes associated with senescence of cardiomyocyte cell lines and results in cardiac aging-associated phenotypes including cardiac remodeling and dysfunction in vivo. Moreover, PQ inhibits the activation of Forkhead box O3 (FoxO3), an important longevity factor, both in vitro and in vivo. We found that PQ-induced senescence phenotypes, including proliferation inhibition, apoptosis, senescence-associated ß-galactosidase activity, and p16INK4a expression, were significantly enhanced by FoxO3 deficiency in cardiomyocytes. Notably, PQ-induced cardiac remolding, apoptosis, oxidative damage, and p16INK4a expression in hearts were exacerbated by FoxO3 deficiency. In addition, both in vitro deficiency and in vivo deficiency of FoxO3 greatly suppressed the activation of antioxidant enzymes including catalase (CAT) and superoxide dismutase 2 (SOD2) in the presence of PQ, which was accompanied by attenuation in cardiac function. The direct in vivo binding of FoxO3 to the promoters of the Cat and Sod2 genes in the heart was verified by chromatin immunoprecipitation (ChIP). Functionally, overexpression of Cat or Sod2 alleviated the PQ-induced senescence phenotypes in FoxO3-deficient cardiomyocyte cell lines. Overexpression of FoxO3 and CAT in hearts greatly suppressed the PQ-induced heart injury and phenotypes associated with aging. Collectively, these results suggest that FoxO3 protects the heart against an aging-associated decline in cardiac function in mice exposed to PQ, at least in part by upregulating the expression of antioxidant enzymes and suppressing oxidative stress.


Subject(s)
Aging/metabolism , Antioxidants/metabolism , Forkhead Box Protein O3/metabolism , Paraquat/antagonists & inhibitors , Protective Agents/metabolism , Up-Regulation , Aging/drug effects , Animals , Catalase/genetics , Catalase/metabolism , Heart/drug effects , Mice , Mice, Knockout , Paraquat/pharmacology , Phenotype , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Up-Regulation/drug effects
5.
Biofactors ; 45(5): 774-787, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31206890

ABSTRACT

Rosmarinic acid (RA) has a wide range of biological effects, including the antioxidation and antiaging. However, the detailed mechanisms remain unclear but highly attractive. Herein, RA promoted lifespan and motoricity in a dose-dependent manner, and reduced fat store without threatening fertility in Caenorhabditis elegans. In term of antioxidant efficacy, catalase activity, glutathione peroxidas activity, reduced glutathione content, and reduced glutathione/oxidized glutathione ratio were enhanced. And malondialdehyde content was diminished significantly. Moreover, RA increased survival under acute oxidative and thermal stress, and suppressed intestinal lipofuscin accumulation. So the improvement of lifespan mediated by RA could be related with its strong antioxidant properties. Furthermore, RA was absorbed by worms. Further research in pursuit of the mechanism showed that longevity induced by RA was involved with the genes sod-3, sod-5, ctl-1, daf-16, ins-18, skn-1, and sek-1, but was independent of subcellular localization of DAF-16. These findings indicated that RA had a potential for promoting healthy lifespan.


Subject(s)
Antioxidants/pharmacology , Caenorhabditis elegans/drug effects , Cinnamates/pharmacology , Depsides/pharmacology , Gene Expression Regulation/drug effects , Longevity/drug effects , MAP Kinase Signaling System/drug effects , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Catalase/genetics , Catalase/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fertility/drug effects , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Glutathione/metabolism , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Lipofuscin/metabolism , Locomotion/drug effects , Longevity/genetics , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Malondialdehyde/metabolism , Oxidative Stress/drug effects , Paraquat/antagonists & inhibitors , Paraquat/pharmacology , Peptide Hormones/genetics , Peptide Hormones/metabolism , Reactive Oxygen Species/agonists , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Rosmarinic Acid
6.
Free Radic Res ; 53(6): 680-693, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31106605

ABSTRACT

Paraquat (PQ) is a widely used agro-chemical in agriculture and highly toxic to humans. Although the mechanism of PQ poisoning is not clear, it has been well documented that reactive oxygen species (ROS) generation and apoptosis play pivotal roles. Alternatively, chlorogenic acid (CA) is a biologically active dietary polyphenol, playing several therapeutic roles. However, it is not known whether CA has protective effect on PQ-induced apoptosis. Here, we investigated the effect of CA in preventing PQ-induced apoptosis and explored the underlying mechanisms. A549 cells were pretreated with 100 µM CA for 24 h and then exposed to 160 µM PQ for 24 h. We found that CA was effective in preventing PQ-induced apoptotic features, including the release of cytochrome c from the mitochondria to cytoplasm, the cleavages of caspase 3 and caspase 9, and the increases in levels of Bcl-2-associated X protein (Bax) and intracellular calcium ions. CA alleviated ROS production and prevented the reduction of antioxidant capacity in cells exposed to PQ by increasing NF-E2-related factor 2 (Nrf2), superoxide dismutase 2 (SOD2) and glutathione levels. In addition, CA also attenuated PQ-induced alterations of mitochondrial structure and function (such as the decreases in membrane potential and adenosine triphosphate level), and the impaired autophagic flux was improved by CA. Down-regulation of sirtuin 1 (Sirt1) by short hairpin RNA reversed the protective effects of CA. Thus, CA may be viewed as a potential drug to treat PQ-induced lung epithelial cell apoptosis and other disorders with similar pathologic mechanisms.


Subject(s)
Apoptosis/drug effects , Chlorogenic Acid/pharmacology , Mitochondria/drug effects , Paraquat/antagonists & inhibitors , Sirtuin 1/metabolism , A549 Cells , Cell Survival/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Homeostasis/drug effects , Humans , Mitochondria/metabolism , Oxidation-Reduction/drug effects , Paraquat/pharmacology , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured
7.
Free Radic Res ; 53(5): 562-573, 2019 May.
Article in English | MEDLINE | ID: mdl-31039619

ABSTRACT

The main flavonoid components of Radix Tetrastigma (RTF) were extracted and identified by UPLC-TOF/MS. In vitro, RTF prevented inflammation in RAW 264.7 cells by suppressing morphological (both cell and nucleus) changes, and decreasing nitric oxide (NO), inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2) contents. Exposure to LPS also leads to oxidant damage, and RTF alleviated damage to mitochondria, decreased O2- accumulation, and restored the glutathione level. RTF intervention decreased the expression of c-Jun N-terminal kinase (JNK) and p38 phosphorylation, accompanied by downregulation of nuclear factor erythroid 2-related factor 2 (Nrf2) and forkhead box protein O1 (FoxO1). In vivo, aging of Caenorhabditis elegans (C. elegans) by paraquat (PQ) was observed through lifespan, lipofuscin, and enzyme analysis. RTF protected against damage in N2 worms but not in daf-16 mutants. Gene expression was further assessed, and p38/PMK-1 and Nrf2/SKN-1 expression in worms was suppressed by PQ, which was reversed by RTF treatment. Together, these results suggested that RTF could help ameliorate inflammation-induced damage through JNK, p38 and Nrf2 pathways.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/drug effects , DNA-Binding Proteins/genetics , Flavonoids/pharmacology , Longevity/drug effects , Mitogen-Activated Protein Kinases/genetics , Transcription Factors/genetics , Vitaceae/chemistry , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , DNA-Binding Proteins/metabolism , Flavonoids/chemistry , Flavonoids/isolation & purification , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Gene Expression Regulation , Glutathione/metabolism , Inflammation , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Longevity/genetics , Mice , Mitochondria/drug effects , Mitochondria/pathology , Mitogen-Activated Protein Kinases/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/drug effects , Paraquat/antagonists & inhibitors , Paraquat/pharmacology , Plant Extracts/chemistry , RAW 264.7 Cells , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Transcription Factors/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Phytochemistry ; 163: 11-22, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30974397

ABSTRACT

Plant cells have a variety of defense mechanisms to alleviate the deleterious effects of oxidative stress. The present work elucidated a schematic diagram of the proposed pathway of peanut hairy root tissue treated with different elicitors; paraquat (PQ), methyl jasmonate (MeJA), and cyclodextrin (CD). The different elicitation approaches could provoke intrinsic stress in plant cells and might activate a distinct response pathway, allowing plants to overcome the deleterious effects of oxidative stress. Among all strategies, hairy root culture pretreated with PQ followed by application of MeJA plus CD showed an extensive induction of antioxidant defense mechanisms. The expression of the antioxidant enzyme genes and stilbene-synthesized enzyme genes were up-regulated in accordance with the dramatic increase in the production of stilbene compounds. The non-enzymatic antioxidant substances exhibited a highly enhanced capability. The pathogenesis-related protein (PR) genes were also highly up-regulated. In summary, we demonstrated that the interplay among MeJA plus CD and PQ may activate a complex signaling network to regulate plant defense mechanisms involving the up-regulation of detoxifying enzymes, induction of free-radical scavengers and overexpression of genes associated with plant defense pathways.


Subject(s)
Acetates/pharmacology , Antioxidants/pharmacology , Cyclodextrins/pharmacology , Cyclopentanes/pharmacology , Fabaceae/drug effects , Oxylipins/pharmacology , Paraquat/antagonists & inhibitors , Plant Growth Regulators/pharmacology , Acetates/chemistry , Antioxidants/chemistry , Cyclodextrins/chemistry , Cyclopentanes/chemistry , Oxidative Stress/drug effects , Oxylipins/chemistry , Paraquat/pharmacology , Plant Growth Regulators/chemistry , Plant Roots/drug effects
9.
Inflammation ; 42(4): 1441-1455, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31028577

ABSTRACT

Paraquat (PQ), a widely used potent herbicide, generates superoxide anions and other free radicals, leading to severe toxicity and acute lung injury. PQ induces pulmonary fibrosis through epithelial to mesenchymal transition (EMT) characterized by increased number of myofibroblasts. Time-dependent PQ-induced EMT has been evaluated in present investigation where intracellular ROS levels were significantly enhanced after 24 h of PQ intoxication. Anti-inflammatory effects of curcumin have been studied where alveolar epithelial cells (A549 cells) were incubated with curcumin (30 µΜ) for 1 and 3 h before PQ intoxication (700 µM). Western blot and immunocytochemistry studies revealed that pretreatment of A549 cells with curcumin for 3 h before PQ exposure has maintained E-cadherin expression and inhibited PQ induced α-smooth-muscle actin (α-SMA) expression. Transforming growth factor-ß (TGF-ß) that seems to be involved in PQ-induced EMT was enhanced after PQ intoxication, but curcumin pretreatment has effectively inhibited its expression. Immunostaining studies have shown that curcumin pretreatment has significantly reduced matrix metalloproteinase-9 (MMP-9) expressions, which were elevated after PQ intoxication. These results demonstrate that curcumin can regulate PQ-induced EMT by regulating the expression of TGF-ß.


Subject(s)
Curcumin/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Paraquat/pharmacology , Transforming Growth Factor beta/drug effects , A549 Cells , Cadherins/metabolism , Drug Antagonism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Herbicides , Humans , Matrix Metalloproteinase 9/metabolism , Paraquat/antagonists & inhibitors , Transforming Growth Factor beta/metabolism
10.
Theranostics ; 9(3): 633-645, 2019.
Article in English | MEDLINE | ID: mdl-30809298

ABSTRACT

Accidental or suicidal ingestion of the world's most widely used herbicide, paraquat (PQ), may result in rapid multi-organ failure with a 60% fatality rate due to the absence of an effective detoxification solution. Effective, specific antidotes to PQ poisoning have been highly desired. Methods: The binding constant of PQ and a synthetic receptor, cucurbit[7]uril (CB[7]), was first determined in various pH environments. The antidotal effects of CB[7] on PQ toxicity were firstly evaluated with in-vitro cell lines. With in-vivo mice models, the pharmacokinetics and the biodistribution of PQ in major organs were determined to evaluate the influence of CB[7] on the oral bioavailability of PQ. Major organs' injuries and overall survival rates of the mice were systemically examined to evaluate the therapeutic efficacy of CB[7] on PQ poisoning. Results: We demonstrate that CB[7] may complex PQ strongly under various conditions and significantly reduce its toxicity in vitro and in vivo. Oral administration of PQ in the presence of CB[7] in a mouse model significantly decreased PQ levels in the plasma and major organs and alleviated major organs' injuries, when compared to those of mice administered with PQ alone. Further studies indicated that oral administration of CB[7] within 2 h post PQ ingestion significantly increased the survival rates and extended the survival time of the mice, in contrast to the ineffective treatment by activated charcoal, which is commonly recommended for PQ decontamination. Conclusion: CB[7] may be used as a specific oral antidote for PQ poisoning by strongly binding with PQ and inhibiting its absorption in the gastrointestinal tracts.


Subject(s)
Antidotes/administration & dosage , Bridged-Ring Compounds/administration & dosage , Herbicides/antagonists & inhibitors , Imidazoles/administration & dosage , Paraquat/antagonists & inhibitors , Poisoning/therapy , Receptors, Artificial/administration & dosage , Administration, Oral , Animal Structures/pathology , Animals , Antidotes/pharmacokinetics , Antidotes/pharmacology , Bridged-Ring Compounds/pharmacokinetics , Bridged-Ring Compounds/pharmacology , Cell Line , Herbicides/toxicity , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , Mice , Paraquat/toxicity , Survival Analysis
11.
Toxicology ; 410: 65-72, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30205152

ABSTRACT

Paraquat (PQ) as an herbicide has been demonstrated to impair dopaminergic (DAergic) neurons and highly correlate with the etiology of Parkinson's disease (PD). WNT/ß-CATENIN signaling is known for the specification and neurogenesis of midbrain DAergic neurons and implicated as a therapeutic target in treating many diseases, such as cancer and degenerative diseases. LGK974, a WNT pathway inhibitor, is currently under clinical trial for patients with malignancies. Since the exact role of WNT/ß-CATENIN signaling in mediating PD is undetermined, LGK974 was used to examine its effect on the PQ-induced cell model of PD. LGK974 attenuated PQ-induced apoptosis and released mitochondrial pro-poptotic molecules in human neuroblastoma SH-SY5Y cell. PQ increased the levels of ß-CATENIN, non-phosphorylated (Ser33/37/Thr41) ß-CATENIN, and phosphorylated glycogen synthase kinase (GSK)-3α/ß. PQ also increased the nuclear translocation of ß-CATENIN, which can be attenuated by LKG974. Furthermore, LGK974 attenuated the PQ-induced release of mitochondrial proapoptotic factors and WNT agonist 1-induced cell death. Taken together, we have shown for the first time that LGK974 mediated through the WNT/ß-CATENIN pathway to prevent PQ-induced cell death.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Parkinson Disease/pathology , Pyrazines/pharmacology , Pyridines/pharmacology , Wnt Signaling Pathway/drug effects , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Cell Line , Cell Line, Tumor , Glycogen Synthase Kinase 3/biosynthesis , Humans , Immunohistochemistry , Paraquat/antagonists & inhibitors , Paraquat/toxicity , beta Catenin/biosynthesis
12.
Toxicol Appl Pharmacol ; 355: 60-67, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29944852

ABSTRACT

A high incidence of intentional or accidental paraquat (PQ) ingestion is related to irreversible lung fibrosis and no effective therapy is currently available. Vitamin D has emerged with promising results as an immunomodulatory molecule when abrogating the inflammatory responses of lung diseases. Therefore, we have investigated the role of vitamin D treatments on PQ-induced lung fibrosis in male C57/BL6 mice. Lung fibrosis was induced by a single injection of PQ (10 mg/kg; i.p.). The control group received PQ vehicle. Seven days later, after the PQ injection or the vehicle injection, the mice received vitamin D (5 µg/kg, i.p., once a day) or vehicle, for a further 7 days. Twenty-four hours after the last dose of vitamin D or the vehicle, the analysis were performed. The vitamin D treatments reduced the number of leukocytes in their BALF and they decreased the IL-6, IL-17, TGF-beta and MMP-9 levels and the abrogated collagenase deposits in their lung tissues. Conversely, the vitamin D treatments increased the resolvin D levels in their BALF. Moreover, their tracheal contractility was also significantly reduced by the vitamin D treatments. Altogether, the data that was obtained showed a promising use of vitamin D, in treating the lung fibrosis that had been induced by the PQ intoxications. This may improve its prognostic use for a non-invasive and low cost therapy.


Subject(s)
Herbicides/toxicity , Inflammation/prevention & control , Paraquat/antagonists & inhibitors , Paraquat/toxicity , Pulmonary Edema/prevention & control , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/prevention & control , Vitamin D/therapeutic use , Vitamins/therapeutic use , Acute Lung Injury , Animals , Bronchoalveolar Lavage Fluid/cytology , Collagen/biosynthesis , Cytokines/metabolism , Inflammation/chemically induced , Leukocyte Count , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle Contraction/drug effects
13.
Sci Rep ; 8(1): 7229, 2018 05 08.
Article in English | MEDLINE | ID: mdl-29739985

ABSTRACT

Apples are well known to have various benefits for the human body. Procyanidins are a class of polyphenols found in apples that have demonstrated effects on the circulatory system and skeletal organs. Osteoarthritis (OA) is a locomotive syndrome that is histologically characterized by cartilage degeneration associated with the impairment of proteoglycan homeostasis in chondrocytes. However, no useful therapy for cartilage degeneration has been developed to date. In the present study, we detected beneficial effects of apple polyphenols or their procyanidins on cartilage homeostasis. An in vitro assay revealed that apple polyphenols increased the activities of mitochondrial dehydrogenases associated with an increased copy number of mitochondrial DNA as well as the gene expression of peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC-1α), suggesting the promotion of PGC-1α-mediated mitochondrial biogenesis. Apple  procyanidins also enhanced proteoglycan biosynthesis with aggrecan upregulation in primary chondrocytes. Of note, oral treatment with apple procyanidins prevented articular cartilage degradation in OA model mice induced by mitochondrial dysfunction in chondrocytes. Our findings suggest that apple procyanidins are promising food components that inhibit OA progression by promoting mitochondrial biogenesis and proteoglycan homeostasis in chondrocytes.


Subject(s)
Cartilage, Articular/drug effects , Chondrocytes/drug effects , Osteoarthritis/prevention & control , Polyphenols/pharmacology , Proanthocyanidins/pharmacology , Proteoglycans/biosynthesis , Animals , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cell Line , Chondrocytes/metabolism , Chondrocytes/pathology , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Disease Models, Animal , Gene Expression , Humans , Male , Malus/chemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Organelle Biogenesis , Osteoarthritis/genetics , Osteoarthritis/metabolism , Osteoarthritis/pathology , Paraquat/antagonists & inhibitors , Paraquat/toxicity , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Polyphenols/isolation & purification , Primary Cell Culture , Proanthocyanidins/isolation & purification , Proteoglycans/agonists , Proteoglycans/genetics , Superoxide Dismutase/deficiency , Superoxide Dismutase/genetics
14.
Free Radic Biol Med ; 110: 133-141, 2017 09.
Article in English | MEDLINE | ID: mdl-28571752

ABSTRACT

Organic selenium and tellurium compounds are known for their broad-spectrum effects in a variety of experimental disease models. However, these compounds commonly display high toxicity and the molecular mechanisms underlying these deleterious effects have yet to be elucidated. Thus, the need for an animal model that is inexpensive, amenable to high-throughput analyses, and feasible for molecular studies is highly desirable to improve organochalcogen pharmacological and toxicological characterization. Herein, we use Caenorhabdtis elegans (C. elegans) as a model for the assessment of pharmacological and toxicological parameters following exposure to two 4-phenylchalcogenil-7-chloroquinolines derivatives (PSQ for selenium and PTQ for tellurium-containing compounds). While non-lethal concentrations (NLC) of PTQ and PSQ attenuated paraquat-induced effects on survival, lifespan and oxidative stress parameters, lethal concentrations (LC) of PTQ and PSQ alone are able to impair these parameters in C. elegans. We also demonstrate that DAF-16/FOXO and SKN-1/Nrf2 transcription factors underlie the mechanism of action of these compounds, as their targets sod-3, gst-4 and gcs-1 were modulated following exposures in a daf-16- and skn-1-dependent manner. Finally, in accordance with a disturbed thiol metabolism in both LC and NLC, we found higher sensitivity of trxr-1 worm mutants (lacking the selenoprotein thioredoxin reductase 1) when exposed to PSQ. Finally, our study suggests new targets for the investigation of organochalcogen pharmacological effects, reinforcing the use of C. elegans as a powerful platform for preclinical approaches.


Subject(s)
Antioxidants/pharmacology , Caenorhabditis elegans/drug effects , Chalcogens/pharmacology , Organometallic Compounds/pharmacology , Organoselenium Compounds/pharmacology , Quinolines/pharmacology , Tellurium/pharmacology , Animals , Antioxidants/chemical synthesis , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Chalcogens/chemical synthesis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Longevity/drug effects , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Organometallic Compounds/chemical synthesis , Organoselenium Compounds/chemical synthesis , Oxidants/antagonists & inhibitors , Oxidants/toxicity , Oxidative Stress , Paraquat/antagonists & inhibitors , Paraquat/toxicity , Quinolines/chemical synthesis , Signal Transduction , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
15.
Exp Mol Med ; 48(10): e264, 2016 10 14.
Article in English | MEDLINE | ID: mdl-27741225

ABSTRACT

This study aimed to validate the high yield and soluble expression of proteins carrying the transactivator of transcription (Tat) peptide tag, and further explored the potential mechanism by which the Tat tag increases expression. Escherichia coli superoxide dismutase (SOD) proteins, including SodA, SodB and SodC, were selected for analysis. As expected, the yields and the solubility of Tat-tagged proteins were higher than those of Tat-free proteins, and similar results were observed for the total SOD enzyme activity. Bacterial cells that overexpressed Tat-tagged proteins exhibited increased anti-paraquat activity compared with those expressing Tat-free proteins that manifested as SodA>SodC>SodB. When compared with an MG1655 wild-type strain, the growth of a ΔSodA mutant strain was found to be inhibited after paraquat treatment; the growth of ΔSodB and ΔSodC mutant strains was also slightly inhibited. The mRNA transcript level of genes encoding Tat-tagged proteins was higher than that of genes encoding Tat-free proteins. Furthermore, the α-helix and turn of Tat-tagged proteins were higher than those of Tat-free proteins, but the ß-sheet and random coil content was lower. These results indicated that the incorporation of the Tat core peptide as a significant basic membrane transduction peptide in fusion proteins could increase mRNA transcripts and promote the high yield and soluble expression of heterologous proteins in E. coli.


Subject(s)
Escherichia coli Proteins/genetics , Escherichia coli/genetics , RNA, Messenger/genetics , Superoxide Dismutase/genetics , Transcriptional Activation , tat Gene Products, Human Immunodeficiency Virus/genetics , Escherichia coli/chemistry , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Gene Expression , HIV-1/genetics , Paraquat/antagonists & inhibitors , Protein Structure, Secondary , Solubility , Superoxide Dismutase/chemistry , Superoxide Dismutase/metabolism , tat Gene Products, Human Immunodeficiency Virus/chemistry , tat Gene Products, Human Immunodeficiency Virus/metabolism
16.
PLoS One ; 11(6): e0157799, 2016.
Article in English | MEDLINE | ID: mdl-27310257

ABSTRACT

Microorganisms produce siderophores to facilitate iron uptake and even though this trait has been extensively studied, there is growing evidence suggesting that siderophores may have other physiological roles aside from iron acquisition. In support of this notion, we previously linked the archetypal siderophore enterobactin with oxidative stress alleviation. To further characterize this association, we studied the sensitivity of Escherichia coli strains lacking different components of the enterobactin system to the classical oxidative stressors hydrogen peroxide and paraquat. We observed that strains impaired in enterobactin production, uptake and hydrolysis were more susceptible to the oxidative damage caused by both compounds than the wild-type strain. In addition, meanwhile iron supplementation had little impact on the sensitivity, the reducing agent ascorbic acid alleviated the oxidative stress and therefore significantly decreased the sensitivity to the stressors. This indicated that the enterobactin-mediated protection is independent of its ability to scavenge iron. Furthermore, enterobactin supplementation conferred resistance to the entE mutant but did not have any protective effect on the fepG and fes mutants. Thus, we inferred that only after enterobactin is hydrolysed by Fes in the cell cytoplasm and iron is released, the free hydroxyl groups are available for radical stabilization. This hypothesis was validated testing the ability of enterobactin to scavenge radicals in vitro. Given the strong connection between enterobactin and oxidative stress, we studied the transcription of the entE gene and the concomitant production of the siderophore in response to such kind of stress. Interestingly, we observed that meanwhile iron represses the expression and production of the siderophore, hydrogen peroxide and paraquat favour these events even if iron is present. Our results support the involvement of enterobactin as part of the oxidative stress response and highlight the existence of a novel regulation mechanism for enterobactin biosynthesis.


Subject(s)
Enterobactin/biosynthesis , Escherichia coli/genetics , Gene Expression Regulation , Siderophores/biosynthesis , Stress, Physiological/genetics , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Carboxylic Ester Hydrolases/genetics , Carboxylic Ester Hydrolases/metabolism , Chlorides/pharmacology , Enterobactin/genetics , Escherichia coli/drug effects , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Ferric Compounds/pharmacology , Hydrogen Peroxide/antagonists & inhibitors , Hydrogen Peroxide/pharmacology , Hydrolysis , Iron/metabolism , Ligases/genetics , Ligases/metabolism , Mutation , Oxidants/antagonists & inhibitors , Oxidants/pharmacology , Oxidation-Reduction , Oxidative Stress , Paraquat/antagonists & inhibitors , Paraquat/pharmacology , Siderophores/genetics , Transcription, Genetic
17.
J Cell Biochem ; 117(4): 872-80, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26358524

ABSTRACT

Nephrotoxicity induced by chemicals such as paraquat (PQ) is a common clinical phenomenon; therefore, searching for drugs with renal protective effect is of a great practical significance. Our previous investigation found that cycloartenyl ferulate (CF) can antagonize the cytotoxic effect of PQ, and recent studies also revealed a variety of bioactivities of CF. However, specific molecular mechanisms underlying the protective effect of CF have not been explored yet. HPLC detection of PQ content indicated that CF reduced PQ accumulation in HK-2 cells and thereby improved cell survival. Western blot results showed that both PQ and CF did not affect the expression of ABCB1; however, while PQ suppressed the expression of ABCC1, CF upregulated ABCC1 expression and thereby reversed the inhibitory effect of PQ on ABCC1 expression. Meanwhile, HK-2 cells did not express ABCG2. When the expression of ABCC1 was knocked down with siRNA, the inhibitory effect of CF on intracellular PQ accumulation was blocked. Further flow cytometric analysis showed that while PQ significantly induced the appearance of sub-G1 apoptotic peak in cells, CF evidently inhibited apoptosis. TUNEL-DAPI double-staining also detected that PQ significantly induced the occurrence of DNA fragmentation in cells, whereas CF effectively inhibited the effect of PQ. Further results showed that ABCC1 siRNA effectively abolished the protective effect of CF on PQ-induced apoptosis. Taken together, these data demonstrated that in HK-2 cells, CF could antagonize PQ-induced toxicity with the involvement of regulatiion of ABCC1 protein expression, which provides a new strategy for treatments of nephrotoxicity.


Subject(s)
Coumaric Acids/pharmacology , Cytotoxins/antagonists & inhibitors , Epithelial Cells/drug effects , Paraquat/antagonists & inhibitors , Protective Agents/pharmacology , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/deficiency , ATP-Binding Cassette Transporters/genetics , Apoptosis/drug effects , Cell Line , Cytotoxins/toxicity , DNA Fragmentation/drug effects , Epithelial Cells/cytology , Epithelial Cells/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation , Humans , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Paraquat/toxicity , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
18.
Nutr Neurosci ; 19(10): 434-446, 2016 Dec.
Article in English | MEDLINE | ID: mdl-25153704

ABSTRACT

OBJECTIVES: Bacopa monnieri (BM), an ayurvedic medicinal plant, has attracted considerable interest owing to its diverse neuropharmacological properties. Epidemiological studies have shown significant correlation between paraquat (PQ) exposure and increased risk for Parkinson's disease in humans. In this study, we examined the propensity of standardized extract of BM to attenuate acute PQ-induced oxidative stress, mitochondrial dysfunctions, and neurotoxicity in the different brain regions of prepubertal mice. METHODS: To test this hypothesis, prepubertal mice provided orally with standardized BM extract (200 mg/kg body weight/day for 4 weeks) were challenged with an acute dose (15 mg/kg body weight, intraperitoneally) of PQ after 3 hours of last dose of extract. Mice were sacrificed after 48 hours of PQ injection, and different brain regions were isolated and subjected to biochemical determinations/quantification of central monoamine (dopamine, DA) levels (by high-performance liquid chromatography). RESULTS: Oral supplementation of BM for 4 weeks resulted in significant reduction in the basal levels of oxidative markers such as reactive oxygen species (ROS), malondialdehyde (MDA), and hydroperoxides (HP) in various brain regions. PQ at the administered dose elicited marked oxidative stress within 48 hours in various brain regions of mice. However, BM prophylaxis significantly improved oxidative homeostasis by restoring PQ-induced ROS, MDA, and HP levels and also by attenuating mitochondrial dysfunction. Interestingly, BM supplementation restored the activities of cholinergic enzymes along with the restoration of striatal DA levels among the PQ-treated mice. DISCUSSION: Based on these findings, we infer that BM prophylaxis renders the brain resistant to PQ-mediated oxidative perturbations and thus may be better exploited as a preventive approach to protect against oxidative-mediated neuronal dysfunctions.


Subject(s)
Bacopa/chemistry , Dietary Supplements , Herbicides/antagonists & inhibitors , Neurotoxicity Syndromes/prevention & control , Oxidative Stress/drug effects , Paraquat/antagonists & inhibitors , Plant Extracts/therapeutic use , Animals , Antioxidants/standards , Antioxidants/therapeutic use , Biomarkers/metabolism , Brain/drug effects , Brain/metabolism , Dietary Supplements/standards , Dopamine/metabolism , Ethnopharmacology , Herbicides/administration & dosage , Herbicides/toxicity , Injections, Intraperitoneal , Lipid Peroxidation/drug effects , Male , Medicine, Ayurvedic , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/standards , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/metabolism , Paraquat/administration & dosage , Paraquat/toxicity , Plant Extracts/standards , Random Allocation
19.
Neurotoxicology ; 52: 230-42, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26705857

ABSTRACT

Evidence suggests that saffron and its major bioactives exhibit significant neuromodulatory effects in various animal models. However, specific data related to their efficacy to attenuate oxidative stress and neurotoxicity in animal models of Parkinson's disease (PD) are limited. Hence, we investigated the neuroprotective efficacy of saffron methanolic extract (SME) and its active constituent, crocin (CR) employing a Drosophila model of parkinsonism. We focussed on attenuation of Rotenone (ROT)-induced locomotor phenotype, oxidative stress, mitochondrial dysfunction and neurotoxicity in this model. SME and CR-enrichment significantly reduced ROT (500µM) induced mortality, rescued the locomotor phenotype and diminished the enhanced levels of oxidative stress markers in head/body regions of flies. The reduced levels of reduced glutathione (GSH) and total thiols (TSH) resulting from ROT exposure were significantly restored with concomitant enhancement of the antioxidant enzymes activities. Further, ROT-induced mitochondrial dysfunctions (MTT reduction, activities of SDH and NADH-Cyt C reductase (complexes I-III) enzymes) were markedly attenuated by SME/CR enrichment. While ROT elevated the activity of acetylcholinesterase (AChE) in head/body regions, both the treatments caused marked diminution of AChE activity and restored the dopamine levels suggesting their effectiveness to mitigate cholinergic function. Interestingly, SME/CR enrichment significantly delayed the onset of locomotor deficits and extended life span of flies among ROT (50µM)-stressed flies. In a satellite study, flies provided with SME/CR prophylaxis exhibited marked resistance to an acute Paraquat (PQ) challenge as evidenced by the lower incidence of lethality and improved locomotor phenotype. Taken together, the neuroprotective effects of saffron and crocin in the fly model may be largely attributable to its antioxidant action. Based on our findings, we propose that saffron may be exploited as a supplementary therapeutic agent in PD and other oxidative stress mediated neurodegenerative conditions.


Subject(s)
Carotenoids/pharmacology , Crocus/chemistry , Disease Models, Animal , Drosophila melanogaster/drug effects , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/drug therapy , Plant Extracts/pharmacology , Acetylcholinesterase/metabolism , Animals , Antioxidants/metabolism , Biomarkers/metabolism , Dopamine/metabolism , Glutathione/metabolism , Hydrogen Peroxide/metabolism , Locomotion/drug effects , Longevity/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Neuroprotective Agents/therapeutic use , Nitric Oxide/metabolism , Oxidative Stress/drug effects , Paraquat/antagonists & inhibitors , Paraquat/toxicity , Parkinsonian Disorders/diet therapy , Parkinsonian Disorders/metabolism , Phytotherapy , Protein Carbonylation/drug effects , Reactive Oxygen Species/metabolism , Rotenone , Sulfhydryl Compounds/metabolism
20.
Int Immunopharmacol ; 29(2): 722-729, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26362205

ABSTRACT

The present study evaluated the protective effect of artificial sweetener neohesperidin dihydrochalcone (NHDC) against paraquat (PQ)-induced acute liver injury in mice. A single dose of PQ (75mg/kg body weight, i.p.) induced acute liver toxicity with the evidences of increased liver damage biomarkers, aspartate transaminase (AST) and alanine transaminase (ALT) activities in serum. Consistently, PQ decreased the antioxidant capacity by reducing glutathione peroxidase (GP-X), glutathione-S-transferase (GST) and catalase (CAT) activities, glutathione (GSH) level and total antioxidant capacity (T-AOC), as well as increasing reactive oxygen species (ROS) and thiobarbituric acid reactive substances (TBARS) levels. Histopathological examination revealed that PQ induced numerous changes in the liver tissues. Immunochemical staining assay indicated the upregulation of cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) expressions. However, NHDC ameliorates PQ-induced hepatic toxicity in mice by reversing these parameters. Additionally, NHDC significantly inhibited PQ-induced nuclear factor-kappa B (NF-κB) expression and mitochondrial-driven apoptotic signaling. TUNEL assay confirmed that PQ-induced apoptosis was relieved by NHDC. In conclusion, these findings suggested that NHDC showed potent antioxidant, anti-inflammatory and anti-apoptotic effects against PQ-induced acute liver damage.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , Chalcones/pharmacology , Chemical and Drug Induced Liver Injury/prevention & control , Hesperidin/analogs & derivatives , Paraquat/toxicity , Sweetening Agents/pharmacology , Alanine Transaminase/blood , Animals , Antioxidants/metabolism , Aspartate Aminotransferases/blood , Chalcones/therapeutic use , Chemical and Drug Induced Liver Injury/pathology , Hesperidin/pharmacology , Hesperidin/therapeutic use , Liver/pathology , Male , Mice , Paraquat/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Signal Transduction , Sweetening Agents/therapeutic use , Thiobarbituric Acid Reactive Substances/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL