Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Hazard Mater ; 404(Pt B): 124050, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33053467

ABSTRACT

Silica nanoparticles (SiO2 NPs) are extensively applied in various field, which increased their health risks to humans. SiO2 NPs were reported to enter into blood through inhalation and meanwhile, the potential use of SiO2 NPs as drug carriers in vivo allows them to present in blood circulation to induce inflammation of vascular endothelial cells which can be closely related with cardiovascular diseases, whilst the intrinsic mechanism has not been well understood. In this study, we found a regulation of signal axis induced by amorphous SiO2 NPs that triggers pro-inflammatory responses in human umbilical vein endothelial cells (HUVECs). HUVECs exposed with SiO2 NPs generate excess amount of reactive oxygen species (ROS) and lactate dehydrogenase (LDH), together with the up-regulation of cell inflammatory factors [interleukin-1 beta (IL-1ß), interleukin-6 (IL-6), tumor necrotic factor-α (TNF-α)] and cell adhesion molecules [intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1)]. In addition, SiO2 NPs were found to promote the translocation and release of high-mobility group box 1 (HMGB1) from nucleus to cytoplasm, which was demonstrated to be regulated by ROS and NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome. Subsequently, toll-like receptor 4 (TLR4) could bind with HMGB1, up-regulate the expression of myeloid differentiation factor 88 (MyD88) and then activate nuclear factor kappa-B (NF-κB) signaling pathway, ultimately induced the inflammatory response of HUVECs. Overall, out results revealed the related signaling pathways of cell inflammation induced by amorphous SiO2 NPs, which provided new insights in understanding SiO2 NPs-induced cytotoxicity and offered safety guidance for further nanomaterial application.


Subject(s)
HMGB1 Protein , Nanoparticles , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammasomes/metabolism , Inflammation/chemically induced , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nanoparticles/toxicity , Signal Transduction , Silicon Dioxide/toxicity , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
2.
Toxicology ; 381: 39-50, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28238930

ABSTRACT

This study is aimed to investigate the inflammation and neurological dysfunction induced by tetrachloro-p-benzoquinone (TCBQ) through Toll-like receptor 4 (TLR4) signaling. We also investigated the protective role of melatonin as an antioxidant and anti-inflammatory agent. In vitro model was established by rat pheochromocytoma PC12 cells, meanwhile, TLR4 wild-type (C57BL/6) and knockout mice (C57BL/10ScNJ TLR4-/-) were used as in vivo model. In vitro study showed TCBQ exposure enhanced the expression of TLR4, myeloid differentiation factor 88 (MyD88) at both transcriptional and post-transcriptional levels. By contrast, melatonin decreased TLR4 and MyD88 expressions. Moreover, our result indicated that melatonin disrupted the formation of TLR4/MyD88/MD2/CD14 complex. In addition, melatonin terminated TCBQ-mediated phosphorylation of c-Jun N-terminal kinase (JNK), p38, and extracellular regulated protein kinase (ERK) signaling and hampered its downstream pro-inflammatory cytokine releases. In vivo result also indicated TLR4 deficiency partially protected against TCBQ-induced morphological and neuropathological changes in mice brain, suggested the role of TLR4. In conclusion, melatonin modulates TCBQ-mediated inflammatory genes through TLR4/MyD88-dependent signaling pathway. Our current study, to the best of our knowledge, is the first time show melatonin not only disrupt the binding of TLR4 and MyD88, but also restricted the formation of TLR4/MD2/CD14 complex, suggesting that melatonin supplementary may represent a valuable therapeutic strategy for inflammatory neurological dysfunction.


Subject(s)
Chloranil/toxicity , Inflammation/drug therapy , Melatonin/pharmacology , Nervous System Diseases/drug therapy , Toll-Like Receptor 4/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation , Inflammation/chemically induced , Inflammation/pathology , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/metabolism , Lymphocyte Antigen 96/genetics , Lymphocyte Antigen 96/metabolism , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Nervous System Diseases/chemically induced , Phosphorylation , Signal Transduction , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Toxicity Tests, Acute
3.
Toxicol Sci ; 153(2): 303-15, 2016 10.
Article in English | MEDLINE | ID: mdl-27413111

ABSTRACT

Tetrachlorobenzoquinone (TCBQ) is a confirmed active metabolite of a well-known environmental pollutant pentachlorophenol (PCP). Unfortunately, there is insufficient knowledge present available on TCBQ's toxicity. Our previous studies indicated that TCBQ induces inflammatory response in vivo and in vitro; however, its exact mechanism needs further investigation. Toll-like receptors (TLRs) play a crucial role in conveying of inflammatory signaling, whilst high-mobility group box 1 (HMGB1) functions as a transcription-enhancing nuclear protein that regulates inflammation. Indeed, this study demonstrated that TCBQ induces the secretion/translocation of HMGB1, which in turn activates its receptors, TLR family gene (especially TLR4) and receptor for advanced glycation end-products (RAGE) expressions. Consistently, the binding affinity of HMGB1 with its receptors also increased. In the case of HMGB1 or TLR4 deficiency, there were decreases in TCBQ-induced neuroinflammatory cytokine production and neuropathological changes, eg, neuronal loss, astrocyte and macrophage cells activation. Moreover, we found the mobilization of TLR4 into lipid rafts occurs in response to TCBQ exposure, lipid rafts disruptors weakened this effect, suggested lipid rafts play an essential role for TLR4-mediated signal transduction and target inflammatory cytokines expressions. In summary, our current findings revealed a previously unknown mechanism of TCBQ-induced neurological inflammation related to HMGB1-TLR4 signaling.


Subject(s)
Benzoquinones/toxicity , HMGB1 Protein/metabolism , Hydrocarbons, Chlorinated/toxicity , Inflammation/chemically induced , Membrane Microdomains/metabolism , Nervous System/pathology , Toll-Like Receptor 4/metabolism , Acetylation , Animals , Cell Nucleus/metabolism , Cytokines/biosynthesis , Cytoplasm/metabolism , Male , Mice , Mice, Inbred C57BL , Nervous System/metabolism , PC12 Cells , Phosphorylation , Protein Transport , Rats , Receptor for Advanced Glycation End Products/metabolism
4.
Toxicology ; 363-364: 48-57, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27393035

ABSTRACT

Our previous studies demonstrated that tetrachlorobenzoquinone (TCBQ), an active metabolite of pentachlorophenol, has effects on the generation of reactive oxygen species (ROS) and oxidative stress in vitro and in vivo. Nuclear factor erythroid-derived 2-like 2 (Nrf2) is a cellular sensor of electrophilic or oxidative stress that regulates the expression of antioxidant enzymes and defensive proteins. We have illustrated that TCBQ activates Nrf2 signaling by promoting the formation of the Kelch-like ECH-associated protein 1 (Keap1) cross-linking dimer and the formation of an ubiquitination switch from Nrf2 to Keap1. The activation of Nrf2 by TCBQ may serve as an adaptive response to a TCBQ-induced oxidative insult. BTB and CNC homolog 1 (Bach1) compete with Nrf2, leading to the negative regulation of the antioxidant response element (ARE). In this report, we propose that TCBQ induces the dynamic inactivation of Bach1. We observed a rapid nuclear efflux of Bach1 and an accumulation of Nrf2 in nuclei upon TCBQ treatment that precedes the binding of Nrf2 with ARE. We found that the nuclear export of Bach1 is dependent on its chromosomal region maintenance 1 (Crm1) interaction and tyrosine phosphorylation. Although TCBQ induces the ubiquitination of Bach1, TCBQ also increases the mRNA and protein levels of Bach1, returning Bach1 to normal levels. Moreover, we found that TCBQ-induced activation of Nrf2 involves c-Jun N-terminal kinase (JNK)-P62 signaling.


Subject(s)
Basic-Leucine Zipper Transcription Factors/physiology , Benzoquinones/pharmacology , Fanconi Anemia Complementation Group Proteins/physiology , Hydrocarbons, Chlorinated/pharmacology , MAP Kinase Signaling System/drug effects , NF-E2-Related Factor 2/physiology , Basic-Leucine Zipper Transcription Factors/drug effects , Blotting, Western , Cycloheximide/pharmacology , Fanconi Anemia Complementation Group Proteins/drug effects , Hep G2 Cells , Humans , MAP Kinase Signaling System/physiology , NF-E2-Related Factor 2/drug effects , Proto-Oncogene Proteins c-myc/physiology , Real-Time Polymerase Chain Reaction , Ubiquitination
5.
Chem Res Toxicol ; 29(7): 1160-71, 2016 07 18.
Article in English | MEDLINE | ID: mdl-27251440

ABSTRACT

Autophagy is a "self-eating" destructive process that eliminates damaged organelles to maintain cellular homeostasis. Polychlorinated biphenyls (PCBs) are one of the most infamous industrial pollutants, which are ubiquitous in nature. In the present study, we found that an active, quinone-type PCB metabolite (PCB29-pQ) treatment causes an autophagic response through mTOR/p70S6k inhibition in HepG2 and MDA-MB-231 cells. Furthermore, our data suggested that PCB29-pQ enhances autophagosome formation through autophagic vacuole (AV) biogenesis, which evokes autophagic flux and induces AV-lysosome colocalization. The inhibition of autophagy enhanced PCB29-pQ-caused cytotoxicity, suggesting that autophagy serves as pro-survival machinery that plays a protective role in the early stage of PCB29-pQ-induced insult. However, higher concentration of PCB29-pQ exposure (>5 µM) caused autophagic cell death, which implied a shift from "pro-survival" to "pro-death" upon autophagic signaling. N-Acetylcysteine suppressed PCB29-pQ-induced autophagy and cytotoxicity, suggesting that ROS plays an important role in the regulation of PCB29-pQ-induced autophagy. Because autophagy shows significant implications in various human diseases and conditions, our current study provides a new mechanism for PCB-associated toxicity.


Subject(s)
Autophagy/drug effects , Polychlorinated Biphenyls/pharmacology , Quinones/pharmacology , Reactive Oxygen Species/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Cell Line, Tumor , Humans , Polychlorinated Biphenyls/chemistry , Quinones/isolation & purification
6.
Chem Res Toxicol ; 29(3): 421-9, 2016 Mar 21.
Article in English | MEDLINE | ID: mdl-26901245

ABSTRACT

Our previous studies suggested that tetrachlorobenzoquinone (TCBQ) elicits pro-inflammatory activities; however, the mechanism of its toxicity toward vascular endothelial cell has not been characterized. Although TCBQ has been shown to stimulate interleukin-1 beta (IL-1ß) expression, it is unknown whether TCBQ regulates post-translational IL-1ß activation. Using human umbilical vein endothelial cells, we discovered that TCBQ not only promotes the expression of NOD-like receptor family, pyrin domain-containing protein 3 (NLRP3) components [composed of NLRP3, adaptor molecule apoptosis-associated speck like protein containing a caspase activation and recruitment domain (ASC), and pro-caspase 1] but also participates in priming the NLRP3 inflammasome. Activation of the NLRP3 inflammasome results in the maturation and release of IL-1ß. Further experiments showed that K(+) efflux, reactive oxygen species (ROS) production, and mitochondrial DNA damage may be involved in NLRP3 inflammasome activation mediated by TCBQ. Moreover, TCBQ downregulates the ubiquitination of NLRP3, further facilitating the activation of the NLRP3 inflammasome. These results suggest that the NLRP3/IL-1ß signaling pathway plays an important role in TCBQ-induced endothelial cell pro-inflammatory responses, which may point to potential therapeutic approaches against TCBQ-mediated toxicity.


Subject(s)
Benzoquinones/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Hydrocarbons, Chlorinated/pharmacology , Inflammasomes/drug effects , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Processing, Post-Translational/drug effects , Benzoquinones/chemistry , Cells, Cultured , Dose-Response Relationship, Drug , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hydrocarbons, Chlorinated/chemistry , Inflammasomes/metabolism , Interleukin-1beta/agonists , Structure-Activity Relationship
7.
Environ Toxicol Pharmacol ; 41: 241-50, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26745386

ABSTRACT

Tetrachlorobenzoquinone (TCBQ) is a joint metabolite of persistent organic pollutants (POPs), hexachlorobenzene (HCB) and pentachlorophenol (PCP). Previous studies have been reported that TCBQ contributes to acute hepatic damage due to its pro-oxidative nature. In the current study, TCBQ showed the highest capacity on the cytotoxicity, ROS formation and inflammatory cytokines release among four compounds, i.e., HCB, PCP, tetrachlorohydroquinone (TCHQ, reduced form of TCBQ) and TCBQ, in PC 12 cells. Further mechanistic study illustrated TCBQ activates nuclear factor-kappa B (NF-κB) signaling. The activation of NF-κB was identified by measuring the protein expressions of inhibitor of nuclear factor kappa-B kinase (IKK) α/ß, p-IKKα/ß, an inhibitor of NF-κB (IκB) α, p-IκBα, NF-κB (p65) and p-p65. The translocation of NF-κB was assessed by Western blotting of p65 in nuclear/cytosolic fractions, electrophoretic mobility shift assay (EMSA) and luciferase reporter gene assay. In addition, TCBQ significantly induced protein and mRNA expressions of inflammatory cytokines and mediators, such as interleukin-1 beta (IL-1ß), IL-6, tumor necrosis factor-alpha (TNF-α), inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2) and the production of nitric oxide (NO) and prostaglandin E2 (PGE2). Pyrrolidine dithiocarbamate (PDTC), a specific NF-κB inhibitor inhibited these effects efficiently, further suggested TCBQ-induced inflammatory responses involve NF-κB signaling. Moreover, antioxidants, i.e., N-acetyl-l-cysteine (NAC), Vitamin E and curcumin, ameliorated TCBQ-induced ROS generation as well as the activation of NF-κB, which implied that ROS serve as the upstream molecule of NF-κB signaling. In summary, TCBQ exhibits a neurotoxic effect by inducing oxidative stress-mediated inflammatory responses via the activation of IKK/IκB/NF-κB pathway in PC12 cells.


Subject(s)
Benzoquinones/toxicity , Cytokines/genetics , Hydrocarbons, Chlorinated/toxicity , Inflammation/genetics , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Animals , Cell Survival/drug effects , Gene Expression Regulation/drug effects , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Inflammation/chemically induced , Inflammation/immunology , NF-kappa B/genetics , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , PC12 Cells , Rats
8.
Chem Res Toxicol ; 28(11): 2160-9, 2015 Nov 16.
Article in English | MEDLINE | ID: mdl-26451628

ABSTRACT

Polychlorinated biphenyls (PCBs) are a group of persistent organic pollutants. The toxic behavior and mechanism of PCBs individuals and congeners have been extensively investigated. However, there is only limited information on their metabolites. Our previous studies have shown that a synthetic PCB metabolite, PCB29-pQ, causes oxidative damage with the evidence of cytotoxicity, genotoxicity, and mitochondrial-derived intrinsic apoptosis. Here, we investigate the effects of PCB29-pQ on DNA damage checkpoint activation, cell cycle arrest, and death receptor-related extrinsic apoptosis in human liver hepatocellular carcinoma HepG2 cells. Our results illustrate that PCB29-pQ increases the S-phase cell population by down-regulating cyclins A/D1/E, cyclin-dependent kinases (CDK 2/4/6), and cell division cycle 25A (CDC25A) and up-regulating p21/p27 protein expressions. PCB29-pQ also induces apoptosis via the up-regulation of Fas/FasL and the activation of caspase 8/3. Moreover, p53 plays a pivotal role in PCB29-pQ-induced cell cycle arrest and apoptosis via the activation of ATM/Chk2 and ATR/Chk1 checkpoints. Cell cycle arrest and apoptotic cell death were attenuated by the pretreatment with antioxidant N-acetyl-cysteine (NAC). Taken together, these results demonstrate that PCB29-pQ induces oxidative stress and promotes p53-dependent DNA damage checkpoint activation, S-phase cycle arrest, and extrinsic apoptosis in HepG2 cells.


Subject(s)
Benzoquinones/toxicity , DNA Damage , Polychlorinated Biphenyls/toxicity , Apoptosis/drug effects , Carcinoma, Hepatocellular/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/metabolism , Cell Survival/drug effects , Checkpoint Kinase 1 , Checkpoint Kinase 2/metabolism , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Protein Kinases/metabolism , Tumor Suppressor Protein p53/metabolism
9.
Free Radic Biol Med ; 89: 522-32, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26453923

ABSTRACT

Fulminant hepatic failure (FHF) is a lethal clinical syndrome characterized by the activation of macrophages and the increased production of inflammatory mediators. The purpose of this study was to investigate the effects of neohesperidin dihydrochalcone (NHDC), a widely-used low caloric artificial sweetener against FHF. An FHF experimental model was established in mice by intraperitoneal injection of D-galactosamine (d-GalN) (400mg/kg)/lipopolysaccharides (LPS) (10 µg/kg). Mice were orally administered NHDC for 6 continuous days and at 1h before d-GalN/LPS administration. RAW264.7 macrophages were used as an in vitro model. Cells were pre-treated with NHDC for 1h before stimulation with LPS (10 µg/ml) for 6h. d-GalN/LPS markedly increased the serum transaminase activities and levels of oxidative and inflammatory markers, which were significantly attenuated by NHDC. Mechanistic analysis indicated that NHDC inhibited LPS-induced myeloid differentiation factor 88 (MyD88) and TIR-containing adapter molecule (TRIF)-dependent signaling. Transient transfection of TLR4 or MyD88 siRNA inhibited the downstream inflammatory signaling. This effect could also be achieved by the pretreatment with NHDC. The fluorescence microscopy and flow cytometry results suggested that NHDC potently inhibited the binding of LPS to TLR4 in RAW264.7 macrophages. In addition, the inhibitory effect of NHDC on LPS-induced translocation of TLR4 into lipid raft domains played an important role in the amelioration of production of downstream pro-inflammatory molecules. Furthermore, the activation of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) by NHDC inhibited TLR4 signaling. In conclusion, our results suggest that NHDC attenuates d-GalN/LPS-induced FHF by inhibiting the TLR4-mediated inflammatory pathway, demonstrating a new application of NHDC as a hepatoprotective agent.


Subject(s)
Antioxidants/pharmacology , Chalcones/pharmacology , Endotoxins/metabolism , Hesperidin/analogs & derivatives , Liver Failure, Acute/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Animals , Blotting, Western , Disease Models, Animal , Down-Regulation , Electrophoretic Mobility Shift Assay , Flow Cytometry , Fluorescent Antibody Technique , Hesperidin/pharmacology , Immunohistochemistry , Liver Failure, Acute/pathology , Membrane Microdomains/metabolism , Mice , Myeloid Differentiation Factor 88/metabolism , Protein Transport/drug effects , RNA, Small Interfering/genetics , Sweetening Agents/pharmacology , Transfection
10.
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
11.
Neurotoxicology ; 49: 149-57, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26112249

ABSTRACT

Although there are few studies suggested PCP exposure induced developmental and behavioral disorders, however, the occurrence of neurotoxicity and PCP has not been firmly established. Tetrachlorobenzoquinone (TCBQ) is a reactive metabolite of environmental pollutant pentachlorophenol (PCP). To the best of our knowledge, there has no information regarding to the neurological toxic effect of TCBQ available. Here, we demonstrated that TCBQ induces cytotoxicity in pheochromocytoma PC12 cell line, and the mode-of-action analysis indicated the involvement of apoptotic signalings, such as the activation of caspase family proteins, the increased expressions of Fas and Fas-associated death domain (FADD), the loss of mitochondrial membrane potential (MMP), the release of cytochrome c (Cyt c) and the cleavage of the caspase substrates poly(ADP-ribose) polymerase (PARP). BI-6C9, a specific BH3-interacting domain death agonist (Bid) inhibitor, repressed TCBQ-induced Bid truncation, along with the activation of caspase 3 and the release of Cyt c, suggested the cross-talk of extrinsic and intrinsic apoptotic signalings. Furthermore, the inhibition of caspase 8 impaired TCBQ-induced the activation of caspase 3, as well as the release of Cyt c and the cleavage of Bid, suggesting caspase 8 acting as the upstream molecule of Bid, and TCBQ-induced apoptosis is initiated via caspase 8, leads to the activation of caspase 9/3 through Bid-mediated amplification loop. Finally, the pretreatment of antioxidant NAC ameliorated Fas, FADD and caspase 8/3 expressions, which illustrated that TCBQ-induced apoptotic signaling is ROS dependent. Taken together, these results indicated that the cleavage of Bid may play an important role in TCBQ-induced neurotoxicity which promotes the cross-talk of extrinsic and intrinsic apoptotic signalings in PC12 cells.


Subject(s)
Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/metabolism , Benzoquinones/pharmacology , Hydrocarbons, Chlorinated/pharmacology , Mutagens/pharmacology , Signal Transduction/drug effects , Analysis of Variance , Animals , Annexin A5/metabolism , Caspase 8/metabolism , Cell Survival/drug effects , Cytosol/drug effects , Dose-Response Relationship, Drug , Fas-Associated Death Domain Protein/metabolism , L-Lactate Dehydrogenase/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , PC12 Cells , Rats , Time Factors
12.
J Agric Food Chem ; 63(22): 5468-75, 2015 Jun 10.
Article in English | MEDLINE | ID: mdl-25978654

ABSTRACT

Neohesperidin dihydrochalcone (NHDC), a sweetener derived from citrus, belongs to the family of bycyclic flavonoids dihydrochalcones. NHDC has been reported to act against CCl4-induced hepatic injury, but its mechanism is still unclear. We first discovered that NHDC showed a strong ability to scavenge free radicals. In addition, NHDC induces the phase II antioxidant enzymes heme oxygenase 1 (HO-1) and NAD(P)H/quinone oxidoreductase 1 (NQO1) through the activation of the nuclear factor (erythroid-derived 2)-like 2 (Nrf2)/antioxidant response element (ARE) signaling. Further assays demonstrated that NHDC induces accumulation of Nrf2 in the nucleus and augmented Nrf2-ARE binding activity. Moreover, NHDC inhibits the ubiquitination of Nrf2 and suggests the modification of Kelch-like ECH-associated protein 1 (Keap1) and the disruption of the Keap1/Nrf2 complex. c-Jun N-terminal kinase (JNK) and p38 but not extracellular signal-regulated protein kinase (ERK) phosphorylations were up-regulated by NHDC treatment. Taken together, NHDC showed its protective antioxidant effect against CCl4-induced oxidative damage via the direct free radical scavenging and indirect Nrf2/ARE signaling pathway.


Subject(s)
Chalcones/administration & dosage , Chemical and Drug Induced Liver Injury/drug therapy , Free Radical Scavengers/administration & dosage , Hesperidin/analogs & derivatives , Animals , Carbon Tetrachloride/adverse effects , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Hesperidin/administration & dosage , Humans , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Liver/drug effects , Liver/metabolism , Mice , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
13.
Chem Res Toxicol ; 28(6): 1326-37, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25950987

ABSTRACT

Organisms are able to respond to environmental insult to maintain cellular homeostasis, which include the activation of a wide range of cellular adaptive responses with tightly controlled mechanisms. The endoplasmic reticulum (ER) is an organelle responsible for protein folding and calcium storage. ER stress leads to the accumulation of unfolded proteins in the ER lumen. To be against or respond to this effect, cells have a comprehensive signaling system, called unfolded protein response (UPR), to restore homeostasis and normal ER function or activate the cell death program. Therefore, it is critical to understand how environmental insult regulates the ingredients of ER stress and UPR signalings. Previously, we have demonstrated that polychlorinated biphenyl (PCB) quinone caused oxidative stress, cytotoxicity, genotoxicity, and apoptosis in HepG2 cells. Here, we investigated the role of a PCB quinone, PCB29-pQ on ER stress, UPR, and calcium release. PCB29-pQ markedly increased the hallmark genes of ER stress, namely, glucose-regulated protein 78 (GRP78), GRP94, and C/EBP homologous protein (CHOP) on both protein and mRNA levels in HepG2 cells. We also confirmed PCB29-pQ induced ER morphological defects by using transmission electron microscopy. Moreover, PCB29-pQ induced intracellular calcium accumulation and calpain activity, which were significantly inhibited by the pretreatment of BAPTA-AM (Ca(2+) chelator). These results were correlated with the outcome that PCB29-pQ induces ER stress-related apoptosis through caspase family gene 12, while salubrinal and Z-ATAD-FMK (a specific inhibitor of caspase 12) partially ameliorated this effect, respectively. N-Acetyl-l-cysteine (NAC) scavenged ROS formation and consequently alleviated PCB29-pQ-induced expression of ER stress-related genes. In conclusion, our result demonstrated for the first time that PCB quinone leads to ROS-dependent induction of ER stress, and UPR and calcium release in HepG2 cells, and the evaluation of the perturbations of ER stress, UPR, and calcium signaling provide further information on the mechanisms of PCB-induced toxicity.


Subject(s)
Benzoquinones/pharmacology , Calcium/metabolism , Endoplasmic Reticulum Stress/drug effects , Polychlorinated Biphenyls/pharmacology , Unfolded Protein Response/drug effects , Apoptosis/drug effects , Benzoquinones/chemistry , Dose-Response Relationship, Drug , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Hep G2 Cells , Humans , Molecular Structure , Polychlorinated Biphenyls/chemistry , Protein Unfolding/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship , Time Factors , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Tumor Cells, Cultured
14.
Toxicol Appl Pharmacol ; 286(1): 10-6, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25818601

ABSTRACT

Our previous studies demonstrated that polychlorinated biphenyl (PCB) quinone induced oxidative DNA damage in HepG2 cells. To promote genomic integrity, DNA damage response (DDR) coordinates cell-cycle transitions, DNA repair and apoptosis. PCB quinone-induced cell cycle arrest and apoptosis have been documented, however, whether PCB quinone insult induce DNA repair signaling is still unknown. In this study, we identified the activation of DDR and corresponding signaling events in HepG2 cells upon the exposure to a synthetic PCB quinone, PCB29-pQ. Our data illustrated that PCB29-pQ induces the phosphorylation of p53, which was mediated by ataxia telangiectasia mutated (ATM) protein kinase. The observed phosphorylated histone H2AX (γ-H2AX) foci and the elevation of 8-hydroxy-2'-deoxyguanosine (8-OHdG) indicated that DDR was stimulated by PCB29-pQ treatment. Additionally, we found PCB29-pQ activates non-homologous end joining (NHEJ), base excision repair (BER) and nucleotide excision repair (NER) signalings. However, these repair pathways are not error-free processes and aberrant repair of DNA damage may cause the potential risk of carcinogenesis and mutagenesis.


Subject(s)
Benzoquinones/pharmacology , DNA Damage , DNA Repair/drug effects , Polychlorinated Biphenyls/pharmacology , 8-Hydroxy-2'-Deoxyguanosine , Ataxia Telangiectasia Mutated Proteins/metabolism , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Hep G2 Cells , Histones/metabolism , Humans , Oxidative Stress/drug effects , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism
15.
Chem Res Toxicol ; 28(4): 765-74, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25742418

ABSTRACT

Tetrachlorobenzoquinone (TCBQ), a metabolite of industrial herbicide pentachlorophenol, showed hepatotoxicity and genotoxicity through reactive oxygen species (ROS) mechanism in vivo and in vitro models. Nuclear factor erythroid-derived 2-like 2 (Nrf2) is a cellular sensor of oxidative or electrophilic stress, which controls the expression of detoxifying enzymes and antioxidant proteins. Using the human hepatoma HepG2 cell line as an in vitro model, we demonstrated a significant induction of Nrf2 but not its negative regulator Kelch-like ECH-associated protein 1 (Keap1), following exposure to TCBQ. Also, our results clearly demonstrated the translocation of cytosolic Nrf2 into the nucleus. After translocation, Nrf2 subsequently binds to the antioxidant response element (ARE), up-regulated heme oxygenase-1 (HO-1), and NADH quinone oxidoreductase subunit 1 (NQO1), which may be considered as an antioxidative response to TCBQ-intoxication. The luciferase reporter assay confirmed the formation of the Nrf2-ARE complex. Furthermore, mechanism studies proposed that TCBQ promoted the formation of the Keap1 cross-linking dimer, a ubiquitination switch from Nrf2 to Keap1 but not the dissociation of the Keap1-Cullin3 (Cul3) complex.


Subject(s)
Benzoquinones/toxicity , Cullin Proteins/metabolism , Hydrocarbons, Chlorinated/toxicity , Intracellular Signaling Peptides and Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Signal Transduction/drug effects , Ubiquitin/metabolism , Dimerization , Hep G2 Cells , Humans , Kelch-Like ECH-Associated Protein 1 , Protein Processing, Post-Translational , Protein Transport , RNA Processing, Post-Transcriptional , Ubiquitination
16.
PLoS One ; 9(9): e107405, 2014.
Article in English | MEDLINE | ID: mdl-25222049

ABSTRACT

Bazhen decoction is a widely used traditional Chinese medicinal decoction, but the scientific validation of its therapeutic potential is lacking. The objective of this study was to investigate corresponding anti-oxidative, anti-inflammatory and anti-apoptosis activities of Bazhen decoction, using acetaminophen-treated mice as a model system. A total of 48 mice were divided into four groups. Group I, negative control, treated with vehicle only. Group II, fed with 500 mg/kg/day Bazhen decoction for 10 continuous days. Group III, received a single dose of 900 mg/kg acetaminophen. Group IV, fed with 500 mg/kg/day Bazhen decoction for 10 continuous days and a single dose of 900 mg/kg acetaminophen 30 min before last Bazhen decoction administration. Bazhen decoction administration significantly decrease acetaminophen-induced serum ALT, AST, ALP, LDH, TNF-α, IL-1ß, ROS, TBARS and protein carbonyl group levels, as well as GSH depletion and loss of MMP. Bazhen decoction restore SOD, CAT, GR and GPx activities and depress the expression of pro-inflammatory factors, such as iNOS, COX-2, TNF-α, NF-κB, IL-1ß and IL-6, respectively. Moreover, Bazhen decoction down-regulate acetaminophen-induced Bax/Bcl-2 ratio, caspase 3, caspase 8 and caspase 9. These results suggest the anti-oxidative, anti-inflammatory and anti-apoptosis properties of Bazhen decoction towards acetaminophen-induced liver injury in mice.


Subject(s)
Acetaminophen/adverse effects , Drugs, Chinese Herbal/therapeutic use , Inflammation/chemically induced , Inflammation/prevention & control , Liver/drug effects , Oxidative Stress/drug effects , Animals , Apoptosis/drug effects , Male , Mice
17.
Toxicol Appl Pharmacol ; 280(2): 305-13, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25123790

ABSTRACT

This study investigated the protective effects of curcumin on tetrachloro-p-benzoquinone (TCBQ)-induced hepatotoxicity in mice. TCBQ-treatment causes significant liver injury (the elevation of serum AST and ALT activities, histopathological changes in liver section including centrilobular necrosis and inflammatory cells), oxidative stress (the elevation of TBAR level and the inhibition of SOD and catalase activities) and inflammation (up-regulation of iNOS, COX-2, IL-1ß, IL-6, TNF-α and NF-κB). However, these changes were alleviated upon pretreatment with curcumin. Interestingly, TCBQ has no effect on caspase family genes or B-cell lymphoma 2 (Bcl-2)/Bcl-2 associated X (Bax) protein expressions, which implied that TCBQ-induced hepatotoxicity is independent of apoptosis. Moreover, curcumin was shown to induce phase II detoxifying/antioxidant enzymes HO-1 and NQO1 through the activation of nuclear factor erythroid-derived 2-like 2 (Nrf2). In summary, the protective mechanisms of curcumin against TCBQ-induced hepatoxicity may be related to the attenuation of oxidative stress, along with the inhibition of inflammatory response via the activation of Nrf2 signaling.


Subject(s)
Apoptosis/drug effects , Benzoquinones/toxicity , Curcumin/toxicity , Hydrocarbons, Chlorinated/toxicity , Inflammation/chemically induced , Liver/drug effects , Oxidative Stress/drug effects , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Caspases/metabolism , Interleukin-6/analysis , Liver/metabolism , Liver/pathology , Male , Mice , NF-E2-Related Factor 2/physiology , NF-kappa B/analysis , Nitric Oxide Synthase Type II/metabolism , Proto-Oncogene Proteins c-bcl-2/analysis
18.
Int Immunopharmacol ; 22(2): 293-302, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25046589

ABSTRACT

This study investigated the protective effect of α-lipoic acid (LA) on lipopolysaccharide (LPS)/d-galactosamine (d-GalN)-induced fulminant hepatic failure in mice. First, we found that LA markedly reduced LPS/d-GalN-induced increases in serum ALT and AST activities, which were supplemented with histopathological examination, suggested that LA has a protective effect on this model of hepatic damage. Livers challenged with LPS/d-GalN exhibited extensive areas of vacuolization with the disappearance of nuclei and the loss of hepatic architecture. On the contrary, these pathological alterations were ameliorated by LA treatment. Next, we found that ROS and TBARS levels were increased in LPS/d-GalN treated liver homogenates, which were attenuated by LA administration. Consistently, decreases in hepatic CAT and GPx activities were observed in LPS/d-GalN group and were significantly restored by LA administration. Moreover, pretreatment with LA markedly reduced LPS/d-GalN-induced iNOS, COX-2, TNF-α, NF-κB, IL-1ß and IL-6 expressions. Furthermore, our data showed that TUNEL-positive cells increased in LPS/d-GalN-treated mice liver which was counteracted by LA administration. LPS/d-GalN induced apoptosis of hepatocytes, as estimated by caspase 3, caspase 8 and caspase 9 activations. Also, the increasing of Bax and the decreasing of Bcl-2 expressions also supported LPS/d-GalN induced apoptosis. Interestingly, LA marked relieved these apoptotic features. Taking together, our results indicated that LA plays an important role on LPS/d-GalN-induced fulminant hepatic failure through its antioxidant, anti-inflammatory and anti-apoptotic activities.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Liver Failure, Acute/drug therapy , Protective Agents/therapeutic use , Thioctic Acid/therapeutic use , Alanine Transaminase/blood , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Aspartate Aminotransferases/blood , Caspases/metabolism , Catalase/metabolism , Cytokines/metabolism , Galactosamine , Glutathione Peroxidase/metabolism , Lipopolysaccharides , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Failure, Acute/chemically induced , Liver Failure, Acute/metabolism , Liver Failure, Acute/pathology , Male , Mice , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/drug effects , Protective Agents/pharmacology , Reactive Oxygen Species/metabolism , Thioctic Acid/pharmacology
19.
Life Sci ; 109(1): 37-43, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-24931906

ABSTRACT

AIMS: This study was designed to investigate the protective effects of selenium supplementation on patulin-induced neurotoxicity. MAIN METHODS: Mice were subjected to patulin for 8 weeks. Sodium selenite (Na2SeO3) and selenium-methionine (Se-Met) were supplemented with the diet, and we investigated the effects of selenium on patulin-induced neurotoxicity. The animals were randomly divided into 4 groups containing 6-8 mice each. The first group was used as a control, and only physiological saline (0.9%) was injected. The second group was treated with patulin (1mg/kg) intraperitoneally. The third group was treated with patulin (1mg/kg) along with a dietary supplementation of Na2SeO3 (0.2mg Se/kg of diet). The fourth group was treated with patulin (1mg/kg) plus Se-Met (0.2mg Se/kg of diet). KEY FINDINGS: Patulin treatment increased oxidative damage in the brain, as evidenced by a decrease in non-protein thiol and total thiol groups, along with significant increases in GSSG, reactive oxygen species, thiobarbituric acid reactive substances and protein carbonyl levels. Moreover, the activities of glutathione peroxidase (GPx) and glutathione reductase were inhibited with patulin treatment. Selenium supplementation significantly ameliorated these biological parameter changes. In addition, selenium treatments significantly increased the mRNA levels of GPx-1, GPx-4 and thioredoxin reductase. SIGNIFICANCE: Our data show that selenium supplementation increases the activity and expression of glutathione-related enzymes and offers significant protection against brain damage induced by patulin.


Subject(s)
Brain/drug effects , Glutathione/metabolism , Mycotoxins/adverse effects , Patulin/adverse effects , Selenomethionine/therapeutic use , Sodium Selenite/therapeutic use , Animals , Brain/metabolism , Brain/pathology , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism , Male , Mice , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Thiobarbituric Acid Reactive Substances/metabolism , Trace Elements/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...