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










Publication year range
1.
Environ Pollut ; 258: 113693, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31838391

ABSTRACT

2,2',4,4'-Tetrabromodiphenyl ether (BDE-47) is associated with various adverse human health effects; however, the knowledge of its toxicity is still very limited. Mitochondrial injury has been observed in liver cells exposed to BDE-47 in vitro. Mitophagy impairment causes the accumulation of dysfunctional mitochondria, contributing to the pathological mechanisms of liver injury. The aim of this study was to investigate whether BDE-47 impairs mitophagy to trigger mitochondrial dysfunction-related liver injury and the underlying mechanisms. This study revealed that BDE-47 elicited mitochondrial dysfunction and related oxidative liver injury by impairing mitophagy. Moreover, our results showed that NAD+ insufficiency is responsible for BDE-47-mediated mitophagy defect and mitochondrial dysfunction in mouse livers, which was associated with suppression of Sirt3/FoxO3a/PINK1 signaling. Furthermore, our results indicated a potential role of miR-34a-5p in the hepatotoxicity of BDE-47. Mechanistically, BDE-47 dramatically upregulated miR-34a-5p expression in mouse livers. The data from AAV-sponge-mediated miR-34a-5p inhibition suggested that miR-34a-5p diminished NAD+ level by directly targeting NAMPT expression in BDE-47-treated mouse livers, which was confirmed by luciferase reporter assay. Consequently, miR-34a-5p markedly abated Sirt3/FoxO3a/PINK1 signaling-mediated mitophagy to promote mitochondrial dysfunction in BDE-47-treated mouse livers. The present study provided in vivo evidence to reveal a potential mechanism for BDE-47-induced mitochondrial dysfunction and related liver injury and indicated that miR-34a-5p-mediated mitophagy impairment might be a therapeutic target for BDE-47 toxicity.


Subject(s)
Chemical and Drug Induced Liver Injury/physiopathology , Halogenated Diphenyl Ethers/toxicity , MicroRNAs/genetics , Mitochondria, Liver/pathology , Mitophagy , Animals , Liver/drug effects , Male , Mice , Mice, Inbred ICR
2.
Mol Med ; 25(1): 29, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31215394

ABSTRACT

BACKGROUND: Innate immune dysfunction contributes to the development and progression of nonalcoholic fatty liver disease (NAFLD), however, its pathogenesis is still incompletely understood. Identifying the key innate immune component responsible for the pathogenesis of NAFLD and clarifying the underlying mechanisms may provide therapeutic targets for NAFLD. Recently, F-box- and WD repeat domain-containing 7 (FBXW7) exhibits a regulatory role in hepatic glucose and lipid metabolism. This study aims to investigate whether FBXW7 controls high-mobility group box 1 protein (HMGB1)-mediated innate immune signaling to improve NAFLD and the mechanism underlying this action. METHODS: Mice were fed a high-fat diet (HFD) for 12 or 20 weeks to establish NAFLD model. Hepatic overexpression or knockdown of FBXW7 was induced by tail-vein injection of recombinant adenovirus. Some Ad-FBXW7-injected mice fed a HFD were injected intraperitoneally with recombinant mouse HMGB1 to confirm the protective role of FBXW7 in NAFLD via inhibition of HMGB1. RESULTS: FBXW7 improves NAFLD and related metabolic parameters without remarkable influence of body weight and food intake. Moreover, FBXW7 markedly ameliorated hepatic inflammation and insulin resistance in the HFD-fed mice. Furthermore, FBXW7 dramatically attenuated the expression and release of HMGB1 in the livers of HFD-fed mice, which is associated with inhibition of protein kinase R (PKR) signaling. Thereby, FBXW7 restrains Toll-like receptor 4 (TLR4) and receptor for advanced glycation end products (RAGE) signaling in HFD-fed mouse livers. In addition, exogenous HMGB1 treatment abolished FBXW7-mediated inhibition of hepatic inflammation and insulin resistance in HFD-fed mouse livers. CONCLUSIONS: Our results demonstrate a protective role of FBXW7 in NAFLD by abating HMGB1-mediated innate immune signaling to suppress inflammation and consequent insulin resistance, suggesting that FBXW7 is a potential target for therapeutic intervention in NAFLD development.


Subject(s)
F-Box-WD Repeat-Containing Protein 7/metabolism , HMGB1 Protein/metabolism , Liver/metabolism , Mice, Inbred C57BL/physiology , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Blotting, Western , F-Box-WD Repeat-Containing Protein 7/genetics , Fluorescent Antibody Technique , Glucose Tolerance Test , HMGB1 Protein/genetics , Immunity, Innate/genetics , Immunohistochemistry , Insulin Resistance/genetics , Insulin Resistance/physiology , Lipid Metabolism/genetics , Lipid Metabolism/physiology , Male , Mice , Mice, Inbred C57BL/genetics , Non-alcoholic Fatty Liver Disease/genetics , Signal Transduction/genetics , Signal Transduction/physiology
3.
Cancer Cell Int ; 19: 48, 2019.
Article in English | MEDLINE | ID: mdl-30867651

ABSTRACT

BACKGROUND: Nasopharyngeal carcinoma (NPC) is a common malignant tumor of the head and neck region with poorly understood progression and prognosis. The present study aims at exploring whether the expression of ß-catenin, TCF-4, and survivin affects clinicopathological features and prognostic significance in NPC. METHODS: We enrolled 164 patients with NPC and 70 patients with chronic nasopharyngitis (CNP) in this study. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) and immunohistochemistry (IHC) were conducted to evaluate the expression of ß-catenin, TCF-4, and survivin. Spearman's rank correlation analysis and Pearson correlation analysis were used to measure the correlation of ß-catenin, TCF-4, and survivin. Risk factors for prognosis and survival conditions of NPC patients were analyzed by Cox proportional hazards model and Kaplan-Meier curves. RESULTS: The results obtained revealed that mRNA and protein expression of ß-catenin, TCF-4, and survivin was higher in NPC tissues than in CNP tissues. Positive correlations amongst ß-catenin, TCF-4, and survivin were identified by Spearman's rank correlation analysis and Pearson correlation analysis. There was a significant correlation in expression of ß-catenin, TCF-4, and survivin with EBV DNA, EBV-VCA-IgA, EBV-EA-IgA, T stage, N stage, and clinicopathological stages. Lower overall survival (OS), distant metastasis-free survival (DMFS), local recurrence-free survival (LRFS), and disease-free survival (DFS) rates were detected in NPC patients with positive expression of ß-catenin, TCF-4, and survivin, in contrast to those with negative expression. Cox proportional hazards model demonstrated that ß-catenin, TCF-4, and survivin protein positive expression were independent risk factors for OS and DFS of NPC prognosis; there was an evident correlation between clinicopathological stages, TCF-4, and EBV-EA-IgA and OS, DMFS, LRFS, and DFS of NPC. CONCLUSIONS: The aforementioned results indicate that ß-catenin, TCF-4, and survivin proteins are highly expressed in NPC, which can be used as factors to predict the malignancy of NPC. In addition, positive expression of ß-catenin, TCF-4, and survivin are potential risk factors that lead to an unfavorable prognosis of OS and DFS in NPC patients.

4.
Oxid Med Cell Longev ; 2019: 5189819, 2019.
Article in English | MEDLINE | ID: mdl-30805082

ABSTRACT

Our preliminary data showed that VEGFR2 upregulation promoted renal ROS overproduction in high-fat diet- (HFD-) treated mice. Given that ROS-induced NLRP3 activation plays a central role in the pathogenesis of type 2 diabetic kidney injury, we evaluate whether VEGFR2 upregulation induces type 2 diabetic kidney injury via ROS-mediated NLRP3 activation and further explore the underlying mechanism. Our results showed that VEGFR2 knockdown decreased ROS overproduction, blocked NLRP3-dependent inflammation, and alleviated kidney damage in HFD-treated mice. Treatment with α-lipoic acid, a scavenger of ROS, lowered ROS overproduction and alleviated NLRP3-triggered kidney injury of HFD-treated mice. Collectively, the VEGFR2/ROS/NLRP3 signal is a critical therapeutic strategy for the kidney injury of HFD-treated mice. Purple sweet potato color (PSPC), a natural anthocyanin, can exert renal protection by inhibiting ROS in HFD-treated mice. Here, we provide a novel mechanism of PSPC against renal damage in HFD-treated mice by downregulating VEGFR2 expression.


Subject(s)
Ipomoea batatas/chemistry , Kidney/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pigments, Biological/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Diet, High-Fat , Gene Knockdown Techniques , Inflammation/pathology , Kidney/drug effects , Male , Mice, Inbred ICR , Organ Specificity , Oxidative Stress/drug effects , Pigments, Biological/administration & dosage , Thioctic Acid/pharmacology
5.
Oxid Med Cell Longev ; 2018: 9865495, 2018.
Article in English | MEDLINE | ID: mdl-29849929

ABSTRACT

2,2',4,4'-Tetrabromodiphenyl ether (BDE-47) induces oxidative stress in kidney cells, but the underlying mechanism remains poorly understood. Troxerutin, a natural flavonoid, has potential antioxidant and anti-inflammatory efficacy. In this study, we assessed the effect of troxerutin on kidney damage caused by BDE-47 and investigated the underlying mechanism. The results showed troxerutin reduced reactive oxygen species (ROS) level and urine albumin-to-creatinine ratio (ACR), decreased the activities of inflammatory factors including cyclooxygenase-2 (COX-2), induced nitric oxide synthase (iNOS) and nuclear factor kappa B (NF-κB) in the kidney tissues of BDE-47-treated mice. Furthermore, troxerutin significantly weakened the expression of kidney NLRP3 inflammasome containing NLRP3, ASC, and caspase-1, contributing to the decline of IL-1ß. Additionally, troxerutin inhibited the increased protein level of stromal-derived factor-1(SDF-1), C-X-C chemokine ligand 12 receptor 4 (CXCR4), and thioredoxin interaction protein (TXNIP) caused by BDE-47. Specifically, the immunoprecipitation assay indicated that there was a direct interaction between CXCR4 and TXNIP. CXCR4 siRNA and TXNIP siRNA also decreased the inflammatory damage, which was similar to the action of troxerutin. Our data demonstrated that troxerutin regulated the inflammatory lesions via CXCR4-TXNIP/NLRP3 inflammasome in the kidney of mice induced by BDE-47.


Subject(s)
Halogenated Diphenyl Ethers/toxicity , Hydroxyethylrutoside/analogs & derivatives , Kidney/drug effects , Protective Agents/pharmacology , Signal Transduction/drug effects , Albumins/analysis , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/metabolism , Chemokine CXCL12/metabolism , Creatinine/urine , Cyclooxygenase 2/metabolism , Hydroxyethylrutoside/pharmacology , Kidney/metabolism , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Thioredoxins/antagonists & inhibitors , Thioredoxins/genetics , Thioredoxins/metabolism
6.
J Thorac Dis ; 10(1): 202-211, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29600050

ABSTRACT

BACKGROUND: Allergic asthma is a complex genetic disorder that involves interactions between genetic and environmental factors. Usage of PTEN may be a good therapeutic strategy for the management of allergic inflammation. Thus, the present study aims to explore the effects of phosphatase and tensin homolog (PTEN) gene silencing on airway remodeling and proliferation of airway smooth muscle cells (ASMCs) in a mouse model of allergic asthma. METHODS: A total of 56 healthy female BABL/c mice (weighing between 16 to 22 grams) were selected and were assigned on random into ovalbumin (OVA; mice were stimulated with OVA to induce allergic asthma), OVA + si-PTEN, normal saline (NS; mice were treated with normal saline) and NS + si-PTEN groups. Masson staining was employed in order to observe lung tissue sections. Immunohistochemical staining was used to detect the expression of α-SMA+. Gene silencing was conducted in the NS + si-PTEN and OVA + si-PTEN groups. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) and western blotting were used to detect the mRNA and protein expressions of PTEN in ASMCs of each group. CCK-8 assay and flow cytometry were performed to determine the cell proliferation rate and cell cycle. RESULTS: Airway remodeling and changes of smooth muscle layer were found in allergic asthmatic mice with thick airway walls. The expression of alpha smooth muscle actin (α-SMA+) was significantly higher in ASMCs of the OVA, OVA + si-PTEN and NS + si-PTEN groups compared with ASMCs of the NS group. The mRNA and protein expressions of PTEN reduced in the OVA, OVA + si-PTEN and NS + si-PTEN groups. The rate of ASMCs proliferation in OVA, OVA + si-PTEN and NS + si-PTEN groups were significantly higher than the NS group. The proportion of ASMCs in S and G2 stages increased, while the number of cells in the G1 stage decreased after PTEN gene silencing. CONCLUSIONS: These results demonstrated that PTEN gene silencing might promote proliferation of ASMCs and airway remodeling in a mouse model of allergic asthma.

7.
Technol Cancer Res Treat ; 17: 1533034618756784, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29466930

ABSTRACT

OBJECTIVE: The aim of this study was to evaluate the mechanisms involved with miRNA-708 and its targeting of bone morphogenetic protein and activin membrane-bound inhibitor in cell proliferation, migration, and apoptosis in mice with melanoma via the Wnt and transforming growth factor ß signaling pathways. METHODS: Sixty mice were recruited of which 40 were subsequently assigned into the experimental group (22 mice were successfully established as melanoma model and 18 mice used in tumor xenograft), and the normal control group consisted of 20 mice. B16 cells were assigned to the normal, blank, and negative control, miR-708 mimics, miR-708 inhibitors, si-BAMBI, and miR-708 inhibitors + si-bone morphogenetic protein and activin membrane-bound inhibitor groups. Western blotting and reverse transcription quantitative polymerase chain reaction were employed to detect the expression levels within the tissues and cell lines. TCF luciferase reporter (TOP-FLASH) or a control vector (FOP-FLASH) was applied to detect the activity of the Wnt signaling pathway. MTT3-(4,5-Dimethylthiazol-2-yl)-2,5 diphenyltetrazolium bromide assay, flow cytometry, scratch test, and Transwell assay were conducted, respectively, for cell proliferation, apoptosis, migration, and invasion, while tumor xenograft procedures were performed on the nude mice recruited for the study. RESULTS: Compared to the normal control group, the model group displayed increased expressions of bone morphogenetic protein and activin membrane-bound inhibitor, Wnt10B, P53, and Bcl-2; TOPflash activity; ß-catenin expression; cell proliferation; migration; and invasion capabilities while decreased expressions of miR-708, vascular endothelial growth factor, Fas, Bax, Caspase-3, and cleaved Caspase-3 and apoptosis rate. Compared to the blank and negative control groups, the miR-708 mimics and small-interfering RNA-bone morphogenetic protein and activin membrane-bound inhibitor groups exhibited decreases expressions of bone morphogenetic protein and activin membrane-bound inhibitor, Wnt10B, P53, and Bcl-2 and decreased proliferation, migration, and invasion capabilities, while increases in the apoptosis rate, expressions of vascular endothelial growth factor, Fas, Bax, Caspase-3, and cleaved Caspase-3; however, downregulated levels of TOPflash activity and ß-catenin expression were recorded. The miR-708 inhibitors group displayed an opposite trend. CONCLUSION: Downregulation of miR-708-targeted bone morphogenetic protein and activin membrane-bound inhibitor inhibits the proliferation and migration of melanoma cells through the activation of the transforming growth factor ß pathway and the suppression of Wnt pathway.


Subject(s)
Gene Expression Regulation, Neoplastic , Melanoma/genetics , Melanoma/metabolism , Membrane Proteins/genetics , MicroRNAs/genetics , Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway , Animals , Apoptosis , Biomarkers , Cell Cycle/genetics , Cell Line , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Models, Animal , Genes, Reporter , Humans , Immunohistochemistry , Male , Melanoma/pathology , Mice , RNA Interference , Xenograft Model Antitumor Assays
8.
J Hazard Mater ; 347: 390-402, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29335220

ABSTRACT

Evidence indicates that oxidative stress is the central pathological feature of 2, 2´, 4, 4´-tetrabromodiphenyl ether (BDE-47)-induced neurotoxicity. Protein kinase C delta (PKCδ), an oxidative stress-sensitive kinase, can be proteolytically cleaved to yield a catalytically active fragment (PKCδ-CF) that is involved in various neurodegenerative disorders. Here, we showed that BDE-47 treatment increased ROS, malondialdehyde, and protein carbonyl levels in the mouse hippocampus. In turn, excessive ROS induced caspase-3-dependent PKCδ activation and stimulated NF-κB p65 nuclear translocation, resulting in inflammation in the mouse hippocampus. These changes caused learning and memory deficits in BDE-47-treated mice. Treatment with Z-DEVD-fmk, a caspase-3 inhibitor, or N-acetyl-L-cysteine, an antioxidant, blocked PKCδ activation and subsequently inhibited inflammation, thereby improving learning and memory deficits in BDE-47-treated mice. Our data further showed that activation of ROS-PKCδ signaling was associated with DJ-1 downregulation, which exerted neuroprotective effects against oxidative stress induced by different neurotoxic agents. Adeno-associated viral vector-mediated DJ-1 overexpression in the hippocampus effectively inhibited excessive ROS production, suppressed caspase-3-dependent PKCδ cleavage, blunted inflammation and ultimately reversed learning and memory deficits in BDE-47-treated mice. Taken together, our results demonstrate that DJ-1 plays a pivotal role in BDE-47-induced neurotoxic effects and learning and memory deficits.


Subject(s)
Dependovirus/genetics , Memory Disorders/therapy , Protein Deglycase DJ-1/genetics , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Behavior, Animal/drug effects , Caspase Inhibitors/pharmacology , Genetic Vectors , Halogenated Diphenyl Ethers/toxicity , Hippocampus/drug effects , Hippocampus/metabolism , Learning/drug effects , Male , Memory Disorders/chemically induced , Memory Disorders/metabolism , Mice, Inbred C57BL , NF-kappa B/metabolism , Oligopeptides/pharmacology , Protein Deglycase DJ-1/metabolism , Protein Kinase C-delta/metabolism , Reactive Oxygen Species/metabolism
9.
J Cell Biochem ; 119(2): 2200-2211, 2018 02.
Article in English | MEDLINE | ID: mdl-28857282

ABSTRACT

Our study was performed to elucidate how SOCS-1/3 silencing suppresses renal interstitial fibrosis (RIF) by alleviating renal tubular damage in rat models affected by hydronephrosis. Male Wistar rats were randomly selected to establish hydronephrosis rat model, after which all rats were classified into normal, model, negative control (NC), siRNA-SOCS-1, siRNA-SOCS-3, and siRNA-SOCS-1 + siRNA-SOCS-3 groups. The levels of urine protein, serum creatinine (Scr), and blood urea nitrogen (BUN) were detected. ELISA was performed to determine levels of cystatin (CysC), ß2-microglobulin (ß2-MG), interleukin (IL)-6, and tumor necrosis factor (TNF)-α. RT-qPCR and Western blotting were used for mRNA and protein expressions of SOCS-1, SOCS-3, α-smooth muscle actin (α-SMA), and transforming Growth Factor (TGF)-ß1. Compared with the normal group, the levels of Scr, BUN, urine protein, NAG, CysC, ß2-MG, IL-6, and TNF-α were increased in other groups, as well as elevated mRNA and protein expressions of SOCS-1, SOCS-3, α-SMA, and TGF-ß1. The siRNA-SOCS-1, siRNA-SOCS-3, and siRNA-SOCS-1 + siRNA-SOCS-3 groups were found with decreased levels of Scr, BUN, urine protein, NAG, CysC, ß2-MG, IL-6, and TNF-α, as well as mRNA and protein expressions of SOCS-1, SOCS-3, α-SMA, and TGF-ß1, including positive rates of SOCS-1 and SOCS-3 proteins in comparison with the model and NC groups. In comparison with the siRNA-SOCS-1 and siRNA-SOCS-3 groups, the siRNA-SOCS-1 + siRNA-SOCS-3 group exhibited decreased levels of Scr, BUN, urine protein, NAG, CysC, ß2-MG, IL-6, and TNF-α. Our study demonstrated that silencing of SOCS-1/3 may suppress RIF by alleviating the renal tubular damage in rat models affected by hydronephrosis.


Subject(s)
Hydronephrosis/genetics , Kidney Tubules/pathology , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/genetics , Animals , Creatinine/blood , Disease Models, Animal , Fibrosis , Gene Silencing , Hydronephrosis/pathology , Kidney Tubules/metabolism , Male , Rats , Rats, Wistar
10.
Cancer Biomark ; 21(3): 711-722, 2018 Feb 14.
Article in English | MEDLINE | ID: mdl-29278883

ABSTRACT

OBJECTIVE: MicroRNAs are widely thought to play a regulatory role in gene expression. Although the more unique microRNA expression profiles have been reported in several tumors, there remains a scarcity of knowledge in relation to microRNA expression profiles in GISTs. During this study, through the alteration in the expression of microRNA-152 (miR-152) in gastrointestinal stromal tumor (GIST) cells, we subsequently evaluated its ability to influence the processes associated with cancer, including proliferation, migration, invasion, and apoptosis, as well as the associated mechanisms. METHODS: The expression of miR-152 and cathepsin L (CTSL) in GIST cell lines (GIST882, GIST430, GIST48 and GIST-T1) and normal gastric mucosal cell line RGM-1 were determined. A series of miR-152 mimics, miR-152 inhibitors, and siRNA against CTSL were introduced to treat GIST-T1 cells with the lowest miR-152 and the highest CTSL were assessed. Cell viability, cell cycle entry, apoptosis, and cell migration/invasion were all evaluated by means of CCK-8 assay, flow cytometry analyses of Annexin V-FITC/PI staining, and transwell assays. RESULTS: The target prediction program and luciferase reporter gene assay verified CTSL is the target of miR-152. Regarding the biological significance of miR-152, siRNA knockdown and ectopic expression studies revealed that miR-152 mimic or siRNA against CTSL exposure reduced cell viability and migration/invasion, which resulted in more cells arrested at the S stage, and induced apoptosis. MiR-152 inhibitor exposure was observed to have induced effects on CTSL cells as opposed to those induced by that of the miR-152 mimics. In contrast, miR-152 downregulation abrogated the effects induced by siRNA against CTSL treatment. CONCLUSION: The key findings of this study provided evidence suggesting that miR-152 functions by means of binding to CTSL to induce GIST cell apoptosis and inhibit proliferation, migration, and invasion. The anti-tumor role of miR-152 makes it an attractive therapeutic target for GIST.


Subject(s)
Apoptosis/genetics , Cathepsin L/genetics , Gastrointestinal Stromal Tumors/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , RNA Interference , Cell Cycle/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Cell Survival/genetics , Humans
11.
J Cell Biochem ; 119(4): 3149-3161, 2018 04.
Article in English | MEDLINE | ID: mdl-29073721

ABSTRACT

This study aims to explore whether microRNA-381 (miR-381) mediating CXCR4 affects the renal tubular epithelial cells (RTEC) of renal ischemia reperfusion (I/R) injury. Forty-eight rats were assigned into the I/R (n = 24, successfully established as I/R model) and sham (n = 24) groups. After collecting kidney tissues, immunohistochemistry, and microvascular density (MVD) counting were conducted for CXCR4 positive expression and MVD numbers. RTECs were assigned into the sham, blank, negative control (NC), miR-381 mimics, miR-381 inhibitor, si-CXCR4, and miR-381 inhibitor + si-CXCR4 groups. RT-qPCR and Western blotting were performed for relative expressions in tissues and cells. Cell proliferation and apoptosis were measured by MTT assay and flow cytometry. Results showed that compared with the sham group, positive expression of CXCR4 and MVD number were higher in the I/R group, which exhibited decreased miR-381 and increased expression of CXCR4, stromal cell-derived factor-1 (SDF1), vascular endothelial growth factor (VEGF), hypoxia-inducible factor 1 (HIF-1α) and Tie-2. Dual luciferase reporter gene assay verified that CXCR4 is a target gene of miR-381. MiR-381 expression was lower in the miR-381 inhibitor + si-CXCR4 and miR-381 inhibitor groups and higher in the miR-381 mimics group than the blank and NC groups. Compared with the blank and NC groups, the miR-381 mimics and si-CXCR4 groups exhibited higher cell proliferation but lower cell apoptosis and expression of CXCR4, SDF1, VEGF, HIF-1α, and Tie-2, whereas the miR-381 inhibitor group exhibited the opposite trend. In conclusion, miR-381 may promote RTEC proliferation in rats with renal I/R injury by down-regulating CXCR4.


Subject(s)
Acute Kidney Injury/genetics , Down-Regulation , Kidney Tubules/cytology , MicroRNAs/genetics , Receptors, CXCR4/genetics , Reperfusion Injury/genetics , Acute Kidney Injury/metabolism , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Epithelial Cells/cytology , Kidney Diseases , Male , Rats , Receptors, CXCR4/metabolism , Reperfusion Injury/metabolism
12.
Molecules ; 22(8)2017 Aug 08.
Article in English | MEDLINE | ID: mdl-28786950

ABSTRACT

Purple sweet potato color (PSPC), a class of naturally occurring anthocyanins, exhibits beneficial effects on metabolic syndrome. Sustained inflammation plays a crucial role in the pathogenesis of metabolic syndrome. Here we explored the effects of PSPC on high-fat diet (HFD)-induced hepatic inflammation and the mechanisms underlying these effects. Mice were divided into four groups: Control group, HFD group, HFD + PSPC group, and PSPC group. PSPC was administered by daily oral gavage at doses of 700 mg/kg/day for 20 weeks. Nicotinamide riboside (NR) was used to increase NAD⁺ levels. Our results showed that PSPC effectively ameliorated obesity and liver injuries in HFD-fed mice. Moreover, PSPC notably blocked hepatic oxidative stress in HFD-treated mice. Furthermore, PSPC dramatically restored NAD⁺ level to abate endoplasmic reticulum stress (ER stress) in HFD-treated mouse livers, which was confirmed by NR treatment. Consequently, PSPC remarkably suppressed the nuclear factor-κB (NF-κB) p65 nuclear translocation and nucleotide oligomerization domain protein1/2 (NOD1/2) signaling in HFD-treated mouse livers. Thereby, PSPC markedly diminished the NLR family, pyrin domain containing 3 (NLRP3) inflammasome activation, ultimately lowering the expressions of inflammation-related genes in HFD-treated mouse livers. In summary, PSPC protected against HFD-induced hepatic inflammation by boosting NAD⁺ level to inhibit NLRP3 inflammasome activation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Hepatitis, Animal/drug therapy , Hepatitis, Animal/metabolism , Inflammasomes/metabolism , Ipomoea batatas/chemistry , NAD/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pigments, Biological/pharmacology , Plant Extracts/pharmacology , Animals , Anthocyanins/chemistry , Anthocyanins/pharmacology , Anti-Inflammatory Agents/chemistry , Diet, High-Fat , Endoplasmic Reticulum Stress , Gene Expression Regulation/drug effects , Hepatitis, Animal/pathology , Liver/drug effects , Liver/metabolism , Liver/pathology , Mice , NF-kappa B/metabolism , Nod Signaling Adaptor Proteins/genetics , Nod Signaling Adaptor Proteins/metabolism , Obesity/drug therapy , Obesity/metabolism , Obesity/pathology , Oxidative Stress/drug effects , Pigments, Biological/chemistry , Plant Extracts/chemistry , Protein Transport
13.
Appl Physiol Nutr Metab ; 42(10): 1082-1091, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28636830

ABSTRACT

Our previous work showed that purple sweet potato colour (PSPC), a class of naturally occurring anthocyanins, effectively improved hepatic glucose metabolic dysfunction in high-fat-diet (HFD)-treated mice. This study investigated the effects of PSPC on HFD-induced hepatic steatosis and the signalling events associated with these effects. Mice were divided into 4 groups: control group, HFD group, HFD+PSPC group, and PSPC group. PSPC was administered daily for 20 weeks at oral doses of 700 mg/(kg·day)-1). Our results showed that PSPC significantly improved obesity and related metabolic parameters, as well as liver injury in HFD-treated mice. Moreover, PSPC dramatically attenuated hepatic steatosis in HFD-treated mice. PSPC markedly prevented oxidative stress-mediated Src activation in HFD-treated mouse livers. Furthermore, PSPC feeding remarkably suppressed mitogen-activated protein kinase kinase/extracellular-signal-regulated kinase (MEK/ERK) signalling and consequent CCAAT/enhancer binding protein ß (C/EBPß) activation and restored AMPK activation in HFD-treated mouse livers, which was confirmed by U0126 treatment. Ultimately, PSPC feeding dramatically reduced protein expression of FAS and CD36 and the activation of ACC, and increased the protein expression of CPT1A in the livers of HFD-treated mice, indicating decreased lipogenesis and fatty acid uptake and enhanced fatty acid oxidation. In conclusion, PSPC exhibited beneficial effects on hepatic steatosis, which were associated with blocking Src and C/EBPß activation.


Subject(s)
Anthocyanins/pharmacology , CCAAT-Enhancer-Binding Protein-beta/metabolism , Diet, High-Fat , Extracellular Signal-Regulated MAP Kinases/metabolism , Ipomoea batatas , Liver/drug effects , Non-alcoholic Fatty Liver Disease/prevention & control , Pigments, Biological/pharmacology , src-Family Kinases/metabolism , AMP-Activated Protein Kinases/metabolism , Acetyl-CoA Carboxylase/metabolism , Animals , Anthocyanins/isolation & purification , CD36 Antigens/metabolism , Disease Models, Animal , Energy Metabolism/drug effects , Enzyme Activation , Ipomoea batatas/chemistry , Liver/enzymology , Liver/pathology , Male , Mice, Inbred ICR , Non-alcoholic Fatty Liver Disease/enzymology , Non-alcoholic Fatty Liver Disease/pathology , Obesity/enzymology , Obesity/pathology , Obesity/prevention & control , Oxidative Stress/drug effects , Phytotherapy , Pigments, Biological/isolation & purification , Plants, Medicinal , Signal Transduction/drug effects , Time Factors , fas Receptor/metabolism
14.
Brain Behav Immun ; 65: 99-110, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28532818

ABSTRACT

It is now commonly known that exposure to polybrominated diphenyl ethers (PBDEs) may cause neurotoxicity and cognitive deficits in children as well as adults, but the underlying mechanisms are still not clear. In the present study, we aimed to elucidate the potential underlying mechanism of 2,2',4,4'-tetrabromodiphenyl ether (BDE-47)-induced neurotoxicity and cognitive impairment. Our results showed that BDE-47-treated mice exhibited impaired cognition and robust upregulation of nuclear TDP-43 in the hippocampus. Hippocampus-specific TDP-43 knockdown attenuated hippocampal apoptosis, restored synaptic protein levels and thus improved cognitive dysfunction in BDE-47-treated mice. Furthermore, our data demonstrated that NLRP3 inflammasome activation played a distinct role in the upregulation of nuclear TDP-43 by downregulating Parkin in the hippocampus of BDE-47-treated mice. Knocking down NLRP3 in the hippocampus or inhibiting caspase 1 activity in BDE-47-treated mice effectively increased Parkin expression in the hippocampus, which decreased the levels of nuclear TDP-43 and ultimately abrogated TDP-43-induced neurotoxic effects. Taken together, our data indicate that TDP-43 upregulation mediated by NLRP3 inflammasome activation via Parkin downregulation in the hippocampus induces cognitive decline in BDE-47-treated mice, and suggest that inhibition of NLRP3 or TDP-43 may be a potential strategy for the prevention or treatment of cognitive impairment in BDE-47-induced neurotoxicity and brain diseases.


Subject(s)
Cognitive Dysfunction/metabolism , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/metabolism , Animals , Apoptosis/drug effects , Cognitive Dysfunction/chemically induced , DNA-Binding Proteins/genetics , Halogenated Diphenyl Ethers/pharmacology , Hippocampus/drug effects , Inflammasomes/metabolism , Inflammasomes/physiology , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Neurotoxicity Syndromes/metabolism , Oxidative Stress/drug effects , Transcriptional Activation , Up-Regulation
15.
Food Chem Toxicol ; 82: 19-26, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25957741

ABSTRACT

Quercetin (QE), a natural flavonoid, has been reported to have many benefits and medicinal properties. However, its protective effects against nickel (Ni) induced injury in liver have not been clarified. The aim of the present study was to investigate the effects of quercetin on hepatic DNA methylation and inflammation in mice exposed to nickel. ICR mice were exposed to nickel sulfate with or without quercetin co-administration for 20 days. Our results showed that quercetin administration significantly inhibited nickel-induced liver injury, which was indicated by diagnostic indicators. In exploring the underlying mechanisms of quercetin action, we found that quercetin decreased total DNA methyltransferases (DNMTs) activity and DNA methylation level of the NF-E2 related factor 2 (Nrf2) DNA in livers of nickel-treated mice. Quercetin also induced Nrf2 nuclear translocation and heme oxygenase-1 (HO-1) activity. Moreover, quercetin decreased production of pro-inflammatory markers including TNF-α, IL-1ß and iNOS. Quercetin significantly inhibited the p38 and signal transducer and activator of transcription 1 (STAT1) activation, which in turn inactivated NF-κB and the inflammatory cytokines in livers of the nickel-treated mice. In conclusion, these results suggested that the inhibition of nickel-induced inflammation by quercetin is associated with its ability to modulate Nrf2/HO-1 and p38/STAT1/NF-κB signaling pathway.


Subject(s)
Inflammation/metabolism , Inflammation/prevention & control , Liver/drug effects , Protective Agents/pharmacology , Quercetin/pharmacology , Animals , Heme Oxygenase-1/metabolism , Liver/metabolism , Liver/pathology , Male , Membrane Proteins/metabolism , Mice, Inbred ICR , NF-E2-Related Factor 2/metabolism , Nickel/toxicity , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Food Chem Toxicol ; 72: 295-302, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25088297

ABSTRACT

Proanthocyanidins (PCs), a class of naturally occurring flavonoids, had been reported to possess a variety of biological activities, including anti-oxidant, anti-tumor and anti-inflammatory. In this study, we examined the protective effect of PCs against lead-induced inflammatory response in the rat brain and explored the potential mechanism of its action. The results showed that PCs administration significantly improved behavioral performance of lead-exposed rats. One of the potential mechanisms was that PCs decreased reactive oxygen species production and increased the total antioxidant capacity in the brains of lead-exposed rats. Furthermore, the results also showed that PCs significantly decreased the levels of tumor necrosis factor-α, interleukin 1ß and cyclooxygenase-2 in the brains of lead-exposed rats. Moreover, PCs significantly decreased the levels of beta amyloid and phosphorylated tau in the brains of lead-treated rats, which in turn inhibited endoplasmic reticulum (ER) stress. PCs also decreased the phosphorylation of protein kinase RNA-like ER kinase, eukaryotic translation initiation factor-2, inositol-requiring protein-1, c-Jun N-terminal kinase, p38 and inhibited nuclear factor-κB nuclear translocation in the brains of lead-exposed rats. In conclusion, these results suggested that PCs could improve cognitive impairments by inhibiting brain oxidative stress and inflammatory response.


Subject(s)
Cognition Disorders/pathology , Endoplasmic Reticulum Stress/drug effects , Lead/toxicity , Neuroprotective Agents/pharmacology , Proanthocyanidins/pharmacology , Administration, Oral , Amyloid beta-Peptides/metabolism , Animals , Antioxidants/pharmacology , Behavior, Animal/drug effects , Cognition Disorders/chemically induced , Cyclooxygenase 2/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Interleukin-1beta/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Lipid Peroxidation/drug effects , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , NF-kappa B/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/metabolism , tau Proteins/metabolism
17.
Aquat Toxicol ; 147: 105-11, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24394944

ABSTRACT

Nickel (Ni) is ubiquitous in the biosphere and is a common component of natural fresh waters. When present in high concentrations, it becomes toxic to aquatic organisms. It is known that Ni toxicity may induce oxidative stress and apoptosis. However, the precise mechanism and the pathways that are activated in fish are still unclear. Thus, this study aimed to assess which apoptotic pathways are triggered by Ni in Carassius auratus liver, the main target of waterborne pollutants. Fish were exposed to 10, 25, 50 and 100mg/L of nickel sulfate for 96 h. Our data showed that Ni exposure caused fish weight loss (by 10-12%) and decreased locomotory activity (by 1-25%). Ni exposure significantly decreased the relative lymphocyte count (by 1-24%) and increased the relative count of monocytes (by 25-111%) and neutrophils (by 10-322%) as compared to controls. Ni induced oxidative stress, as evidenced by increasing of lipid peroxidation level (29-91%) and depleting of the glutathione levels (7-79%) in fish liver. Ni also suppressed the activities of superoxide dismutase (by 39-55%) and glutathione peroxidase (16-24%) and decreased ATP levels (13-51%) in livers. Moreover, liver caspase-3, one of the key executioners of apoptosis, was markedly activated by the Ni exposure. Ni exposure also increased expression levels of phosphorylated Jun N-terminal kinases (JNK) in liver, which in turn activated pro-apoptotic signaling events by breaking the balance between pro-apoptotic and anti-apoptotic Bcl-2 proteins. In conclusion, these results suggested that Ni induced oxidative stress and apoptosis, at least, via the JNK signaling pathway.


Subject(s)
Apoptosis/drug effects , Goldfish/physiology , Liver/drug effects , MAP Kinase Signaling System/drug effects , Nickel/toxicity , Oxidative Stress/drug effects , Water Pollutants, Chemical/toxicity , Adenosine Triphosphate/analysis , Animals , Body Weight/drug effects , Enzyme Activation/drug effects , Enzymes/metabolism , Goldfish/metabolism , Liver/chemistry , Motor Activity/drug effects
18.
J Agric Food Chem ; 61(31): 7630-5, 2013 Aug 07.
Article in English | MEDLINE | ID: mdl-23855546

ABSTRACT

Quercetin (QE), the major bioflavonoid in the human diet, has been reported to have many benefits and medicinal properties. However, its protective effects against lead (Pb)-induced neurotoxicity have not been clarified. The aim of the present study was to investigate the effects of QE on neurotoxicity in mice exposed to Pb. Mice were exposed to lead acetate (20 mg/kg body weight/day) intragastrically with or without QE (15 and 30 mg/kg body weight/day) coadministration for 3 months. The data showed that QE significantly prevented Pb-induced neurotoxicity in a dose-dependent manner. Exploration of the underlying mechanisms of QE action revealed that QE administration decreased Pb contents in blood (13.2, 19.1%) and brain (17.1, 20.0%). QE markedly increased NO production (39.1, 61.1%) and PKA activity (51.0, 57.8%) in brains of Pb-treated mice. Additionally, QE remarkably suppressed Pb-induced oxidative stress in mouse brain. Western blot analysis showed that QE increased the phosphorylations of Akt, CaMKII nNOS, eNOS, and CREB in brains of Pb-treated mice. The results suggest that QE can inhibit Pb-induced neurotoxicity and partly restore PKA, Akt, NOS, CaMKII, and CREB activities.


Subject(s)
Brain/drug effects , Lead/toxicity , Neurons/drug effects , Neuroprotective Agents/pharmacology , Quercetin/pharmacology , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Brain/metabolism , Male , Mice , Neurons/metabolism , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
19.
Environ Toxicol Pharmacol ; 35(3): 502-10, 2013 May.
Article in English | MEDLINE | ID: mdl-23501611

ABSTRACT

Puerarin (PU), a natural flavonoid, has been reported to have many benefits and medicinal properties. The present study aimed to investigate the protective effects of puerarin on neurotoxicity in mice exposed to lead. ICR mice were exposed to lead acetate in the drinking water (500 ppm) with or without puerarin coadministration (100 and 200 mgPU/kg intragastrically once daily) for three months. We found puerarin significantly prevented Pb-induced neurotoxicity in a dose-dependent manner, indicated by behavioral indicators. Puerarin also decreased Pb contents in blood and brain. Puerarin increased activities of acetyl cholinesterase (AChE) and monoamine oxidase (MAO) in brain of Pb-treated mice. Moreover, Pb-induced profound elevation of oxidative stress, as evidenced by increasing of lipid peroxidation level and depleting of total antioxidant capacity in brain, were suppressed by treatment with puerarin. Puerarin markedly increased NO production and PKA activity in brain of Pb-treated mice. Western blot analysis showed that puerarin dramatically increased the expression levels of nNOS, eNOS and phosphor-Akt in brains of Pb-treated mice. In conclusion, these results suggested that puerarin can inhibit Pb-induced neurotoxicity, at least in part, by suppressing oxidative stress, reversing the Pb-induced alterations in transmitters and enzymes and modulating the PKA/Akt/NOS signaling pathway.


Subject(s)
Cognition Disorders/drug therapy , Environmental Pollutants/toxicity , Isoflavones/therapeutic use , Lead/toxicity , Neuroprotective Agents/therapeutic use , Neurotoxicity Syndromes/drug therapy , Acetylcholinesterase/metabolism , Animals , Behavior, Animal/drug effects , Brain/drug effects , Brain/metabolism , Cognition Disorders/chemically induced , Cognition Disorders/metabolism , Cognition Disorders/physiopathology , Cyclic AMP-Dependent Protein Kinases/metabolism , Environmental Pollutants/blood , Environmental Pollutants/pharmacokinetics , Isoflavones/pharmacology , Lead/blood , Lead/pharmacokinetics , Lipid Peroxidation/drug effects , Male , Mice , Mice, Inbred ICR , Monoamine Oxidase/metabolism , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/physiopathology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I/metabolism , Nitric Oxide Synthase Type III/metabolism , Protein Carbonylation/drug effects , Reactive Oxygen Species/metabolism , Thiobarbituric Acid Reactive Substances/metabolism
20.
J Agric Food Chem ; 61(5): 1146-54, 2013 Feb 06.
Article in English | MEDLINE | ID: mdl-23317420

ABSTRACT

Sesamin (Ses), one of the major lignans in sesame seeds and oil, has been reported to have many benefits and medicinal properties. However, its protective effects against nickel (Ni)-induced injury in liver have not been clarified. The aim of the present study was to investigate the effects of sesamin on hepatic oxidative DNA injury and apoptosis in mice exposed to nickel. Kunming mice were exposed to nickel sulfate with or without sesamin coadministration for 20 days. The data showed that sesamin significantly prevented nickel-induced hepatotoxicity in a dose-dependent manner, indicated by both diagnostic indicators of liver damage (serum aminotransferase activities) and histopathological analysis. Moreover, nickel-induced profound elevation of reactive oxygen species (ROS) production and oxidative stress, as evidenced by an increase of the lipid peroxidation level and depletion of the intracellular reduced glutathione (GSH) level in liver, were suppressed by treatment with sesamin. Sesamin also restored the activities of antioxidant enzymes (T-SOD, CAT, and GPx) and decreased 8-hydroxy-2-deoxyguanosine (8-OHdG) levels in nickel-treated mice. Furthermore, a TUNEL assay showed that nickel-induced apoptosis in mouse liver was significantly inhibited by sesamin. Exploration of the underlying mechanisms of sesamin action revealed that activities of caspase-3 were markedly inhibited by the treatment of sesamin in the liver of nickel-treated mice. Sesamin increased expression levels of phosphoinositide-3-kinase (PI3K) and phosphorylated protein kinase B (PBK/Akt) in liver, which in turn inactivated pro-apoptotic signaling events, restoring the balance between pro- and anti-apoptotic Bcl-2 proteins in the liver of nickel-treated mice. In conclusion, these results suggested that the inhibition of nickel-induced apoptosis by sesamin is due at least in part to its antioxidant activity and its ability to modulate the PI3K-Akt signaling pathway.


Subject(s)
Apoptosis/drug effects , DNA Damage/drug effects , Dioxoles/pharmacology , Lignans/pharmacology , Liver/drug effects , Nickel/adverse effects , Oxidative Stress/drug effects , 8-Hydroxy-2'-Deoxyguanosine , Alanine Transaminase/blood , Animals , Antioxidants/pharmacology , Aspartate Aminotransferases/blood , Blotting, Western , Caspase 3/genetics , Caspase 3/metabolism , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/genetics , Deoxyguanosine/metabolism , Glutathione , In Situ Nick-End Labeling , Lipid Peroxidation , Liver/metabolism , Male , Mice , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Reactive Oxygen Species , Signal Transduction , Thiobarbituric Acid Reactive Substances/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...