Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Ecotoxicol Environ Saf ; 270: 115943, 2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-38194811

RESUMO

Nickel oxide nanoparticles (NiONPs) are toxic heavy metal compounds that induce liver fibrosis and metabolic disorders. Current research shows that the intestinal microbiota regulates liver metabolism through the gut-liver axis. However, it is unclear whether NiONPs affect the intestinal microbiota and the relationship between microbiota and liver metabolic disorders. Therefore, in this study, we established liver fibrosis model by administering 0.015, 0.06 and 0.24 mg/mL NiONPs through tracheal instillation twice a week for 9 weeks in rats, then we collected serum and fecal sample for whole metabolomics and metagenomic sequencing. As the result of sequencing, we screened out seven metabolites (beta-D-glucuronide, methylmalonic acid, linoleic acid, phosphotidylcholine, lysophosphatidylinositol, docosapentaenoic acid and progesterone) that related to functional alterations (p < 0.05), and obtained a decrease of probiotics abundances (p < 0.05) as well as a variation of the microbiota enzyme activity (p < 0.05), indicating that NiONPs inhibited the proliferation of probiotics. As the result of correlation analysis, we found a positive correlation between differential metabolites and probiotics, such as lysophosphatidylinositol was positively correlated with Desulfuribacillus, Jeotgallibacillus and Rummeliibacillus (p < 0.05). We also found that differential metabolites had correlations with differential proteins and enzymes of intestinal microbiota, such as glucarate dehydratase, dihydroorotate dehydrogenase and acetyl-CoA carboxylase (p < 0.05). Finally, we screened six metabolic pathways with both differential intestinal microbiota enzymes and metabolites were involved, such as pentose and glucuronate interconversions, and linoleic acid metabolism. In vitro experiments showed that NiONPs increased the transcriptional expression of Col1A1 in LX-2 cells, while reducing the mRNA expression of serine/threonine activators, acetyl coenzyme carboxylase, and lysophosphatidylinositol synthase, and short chain fatty acid sodium butyrate can alleviate these variation trends. The results proved that the intestinal microbiota enzyme systems were associated with serum metabolites, suggesting that the disturbance of intestinal microbiota and reduction of probiotics promoted the occurrence and development of NiONPs-induced liver fibrosis by affecting metabolic pathways.


Assuntos
Microbioma Gastrointestinal , Doenças Metabólicas , Ratos , Animais , Microbioma Gastrointestinal/genética , Ácido Linoleico , Cirrose Hepática/induzido quimicamente , Acetil-CoA Carboxilase
2.
Environ Toxicol ; 39(4): 2374-2389, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38165020

RESUMO

Nickel oxide nanoparticles (NiONPs) are an emerging nanomaterial, which poses a huge threat to the health of workplace population. Nanoparticles induce pulmonary fibrosis, and its mechanisms are associated with noncoding RNAs (ncRNAs). However, ncRNAs and competing endogenous RNA (ceRNA) networks which involved in NiONP-induced pulmonary fibrosis are still unclear. This study aimed to identify ncRNA-related ceRNA networks and investigate the role of the Wnt/ß-catenin pathway in pulmonary fibrosis. Male Wistar rats were intratracheally instilled with 0.015, 0.06, and 0.24 mg/kg NiONPs twice a week for 9 weeks. First, we found there were 93 circularRNAs (circRNAs), 74 microRNAs (miRNAs), 124 long non-coding RNAs (lncRNAs), and 1675 messenger RNAs (mRNAs) differentially expressed through microarray analysis. Second, we constructed ceRNA networks among lncRNAs/circRNAs, miRNAs and mRNAs and identified two ceRNA networks (lncMelttl16/miR-382-5p/Hsd17b7 and circIqch/miR-181d-5p/Stat1) after real time-quantitative polymerase chain reaction (RT-qPCR) validation. Furthermore, based on Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses, ncRNAs were found to be involved in biological processes and signaling pathways related to pulmonary fibrosis. KEGG analysis showed that NiONPs activated the Wnt/ß-catenin pathway in rats. In vitro, HFL1 cells were treated with 0, 50, 100, and 200 µg/mL NiONPs for 24 h. We found that NiONPs induced collagen deposition and Wnt/ß-catenin pathway activation. Moreover, a blockade of Wnt/ß-catenin pathway alleviated NiONP-induced collagen deposition. In conclusion, these observations suggested that ncRNAs were crucial in pulmonary fibrosis development and that the Wnt/ß-catenin pathway mediated the deposition of collagen.


Assuntos
MicroRNAs , Nanopartículas , Níquel , Fibrose Pulmonar , RNA Longo não Codificante , Masculino , Ratos , Animais , beta Catenina/metabolismo , RNA Circular , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/genética , Ratos Wistar , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Mensageiro/metabolismo , Perfilação da Expressão Gênica , Via de Sinalização Wnt/genética , Nanopartículas/toxicidade , Colágeno , Redes Reguladoras de Genes
3.
Toxicol Ind Health ; 40(5): 244-253, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38518383

RESUMO

With the widespread use of manganese dioxide nanoparticles (nano MnO2), health hazards have also emerged. The inflammatory damage of brain tissues could result from nano MnO2, in which the underlying mechanism is still unclear. During this study, we aimed to investigate the role of ROS-mediated p38 MAPK pathway in nano MnO2-induced inflammatory response in BV2 microglial cells. The inflammatory injury model was established by treating BV2 cells with 2.5, 5.0, and 10.0 µg/mL nano MnO2 suspensions for 12 h. Then, the reactive oxygen species (ROS) scavenger (20 nM N-acetylcysteine, NAC) and the p38 MAPK pathway inhibitor (10 µM SB203580) were used to clarify the role of ROS and the p38 MAPK pathway in nano MnO2-induced inflammatory lesions in BV2 cells. The results indicated that nano MnO2 enhanced the expression of pro-inflammatory cytokines IL-1ß and TNF-α, elevated intracellular ROS levels and activated the p38 MAPK pathway in BV2 cells. Controlling intracellular ROS levels with NAC inhibited p38 MAPK pathway activation and attenuated the inflammatory response induced by nano MnO2. Furthermore, inhibition of the p38 MAPK pathway with SB203580 led to a decrease in the production of inflammatory factors (IL-1ß and TNF-α) in BV2 cells. In summary, nano MnO2 can induce inflammatory damage by increasing intracellular ROS levels and further activating the p38 MAPK pathway in BV2 microglial cells.


Assuntos
Compostos de Manganês , Microglia , Óxidos , Proteínas Quinases p38 Ativadas por Mitógeno , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Espécies Reativas de Oxigênio/metabolismo , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Linhagem Celular
4.
Environ Toxicol ; 38(11): 2783-2796, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37528634

RESUMO

Nickel oxide nanoparticles (Nano NiO) have been shown to cause pulmonary fibrosis; But, the underlying epigenetic mechanisms remain poorly understood. In this study, we aimed to investigate the role of lncRNA AP000487.1 in regulating PRKCB DNA methylation and the Toll-like receptor 4 (TLR4)/ Myeloid differentiation primary response 88 (MyD88)/ Nuclear factor kappa-B (NF-κB) pathway in Nano NiO-induced collagen formation. We found that lncRNA AP000487.1 was able to bind to the promoter region of the PRKCB gene by Chromosomal RNA pull-down experiments (Ch-RNA pull-down). Moreover, Nano NiO exposure led to down-regulation of lncRNA AP000487.1 expression and PRKCB DNA methylation, resulting in up-regulation of PRKCB expression, activation of the TLR4/MyD88/NF-κB pathway, and increased collagen formation in BEAS-2B cells. Conversely, overexpression of lncRNA AP000487.1 restored PRKCB expression, reduced its hypomethylation and attenuated TLR4/MyD88/NF-κB pathway activation and collagen formation. Furthermore, treatment with the DNA methylation inhibitor, decitabine, alleviated Nano NiO-induced PRKCB2 expression, TLR4/MyD88/NF-κB pathway activation, and collagen formation. Additionally, using PRKCB2 overexpression plasmid, PRKCB2 siRNA, and PRKCB2 protein inhibitor LY317615 influenced NF-κB pathway activity and collagen formation. Finally, TLR4 inhibitor (TAK-242) restrained Nano NiO-induced MyD88/NF-κB pathway activation and excessive collagen formation. In summary, we demonstrated that the down-regulated lncRNA AP000487.1 could cause PRKCB hypomethylation and increased expression, resulting in NF-κB pathway activation and collagen formation in Nano NiO-induced BEAS-2B cells. This is the first study to reveal the role of lncRNA AP000487.1 in regulating collagen formation in Nano NiO-exposed BEAS-2B cells. Our study identified that lncRNA AP000487.1/PRKCB hypomethylation/NF-κB pathway was a regulatory axis of BEAS-2B cells collagen excessive formation. Our findings indicate that lncRNA AP000487.1 and PRKCB DNA methylation may function as biomarkers or potential targets in response to Nano NiO exposure.

5.
Ecotoxicol Environ Saf ; 242: 113925, 2022 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-35917710

RESUMO

Manganese (Mn) is a common environmental pollutant. Mn exposure can lead to neurodegenerative diseases resembling Parkinson's disease, and has become a major public health concern. However, the mechanism of Mn-induced neurotoxicity in the brain is not clear. Fecal microbiome transplantation (FMT) may alleviate the neurotoxicity of Mn exposure by remodeling the gut microbiota. In this study, MnCl2 (manganese chloride) was administered to mice as in drinking water (Mn: 200 mg/L), and fecal matter from donor mice was administered by oral gavage every other day to the recipient mice. The Mn exposure model (Mn group) and FMT model (Mn+FMT group) were established and analyzed 5 weeks post-exposure. The Wipi1 gene exhibited the most significant increase associated with Mn exposure and Mn+FMT treatment groups based on transcriptome analysis. Combined analysis of transcriptomics and proteomics demonstrated that the apelin signaling pathway is the main pathway affected by FMT during Mn exposure. Immunofluorescence and Western blot showed that the expression of key proteins (Beclin-1, LC-3B, and PINK1) associated with autophagy in the hippocampus was robustly activated in the Mn exposure group, but attenuation was observed in Mn+FMT mice, suggesting a critical role of autophagy in neurotoxicity induced by Mn exposure. Our research provides evidence for the neurotoxic effects of Mn exposure through autophagy activation and provides an underlying mechanism of FMT protection against Mn-induced neurotoxicity through regulation of the apelin signaling pathway.


Assuntos
Transplante de Microbiota Fecal , Síndromes Neurotóxicas , Animais , Apelina , Autofagia , Encéfalo , Manganês/toxicidade , Camundongos , Transdução de Sinais
6.
Ecotoxicol Environ Saf ; 248: 114308, 2022 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-36410144

RESUMO

Nickel oxide nanoparticles (NiONPs) induced liver fibrosis, while its mechanisms associated with transcriptome remained unclear. This study aimed to investigate the roles of differentially expressed (DE) messenger RNAs (mRNAs) and non-coding RNAs (ncRNAs) in NiONPs-induced liver fibrosis, and further confirm whether JNK/c-Jun pathway enriched by the DE RNAs was involved in the regulation of the disease. A liver fibrosis rat model was established by intratracheal perfusion of NiONPs twice a week for 9 weeks. Whole-transcriptome sequencing was applied to obtain expression profiles of mRNAs, long non-coding RNAs (lncRNAs), microRNAs (miRNAs), and circular RNAs (circRNAs) in the model rat and control liver tissues. Comparing the RNA expression profiles of the model and control liver tissues, we identified 324 DE mRNAs, 129 DE lncRNAs, 24 DE miRNAs and 33 DE circRNAs, and the potential interactions among them were revealed by constructing two co-expression networks, including lncRNA-miRNA-mRNA and circRNA-miRNA-mRNA networks. Using RT-qPCR, we verified the sequencing results of some RNAs in the networks and obtained similar expression profiles, indicating our sequencing results were reliable and referable. Through Gene Ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, we predicted the biological functions and signaling pathways potentially related to NiONPs-induced liver fibrosis, such as "positive regulation of JNK cascade", "inflammatory response", "transcription factor binding", and MAPK, Wnt, PI3K-Akt signaling pathways. JNK/c-Jun pathway, a subclass of MAPK signal, was selected for further investigation because it was significantly enriched by fibrosis-related DE genes and activated in animal models. In vitro, we detected the cytotoxicity of NiONPs on LX-2 cells and treated the cells with 5 µg/ml NiONPs for 12 h. The results showed NiONPs induced the up-regulated protein expression of fibrotic factors collagen-1a1 (Col-1a1) and matrix metalloproteinas2 (MMP2) and JNK/c-Jun pathway activation. While these effects were reversed after JNK/c-Jun pathway was blocked by SP600125 (JNK pathway inhibitor), indicating the pathway was involved in NiONPs-induced excessive collagen formation. In conclusion, our results revealed the DE mRNAs and ncRNAs played crucial roles in NiONPs-induced liver fibrosis, and JNK/c-Jun pathway mediated the development of the disease.


Assuntos
MicroRNAs , RNA Longo não Codificante , Ratos , Animais , RNA Mensageiro/genética , RNA Longo não Codificante/genética , RNA Circular/genética , Transcriptoma , Fosfatidilinositol 3-Quinases , Cirrose Hepática/genética , MicroRNAs/genética
7.
Environ Toxicol ; 37(5): 1058-1070, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35006638

RESUMO

The lung inflammatory damage could result from the nickel oxide nanoparticles (NiO NPs), in which the underlying mechanism is still unclear. This article explored the roles of long noncoding RNA maternally expressed gene 3 (lncRNA MEG3) and p38 mitogen activated protein kinases (p38 MAPK) pathway in pulmonary inflammatory injury induced by NiO NPs. Wistar rats were treated with NiO NPs suspensions (0.015, 0.06, and 0.24 mg/kg) by intratracheal instillation twice-weekly for 9 weeks. Meanwhile, A549 cells were treated with NiO NPs suspensions (25, 50, and 100 µg/ml) for 24 h. It can be concluded that the NiO NPs did trigger pulmonary inflammatory damage, which was confirmed by the histopathological examination, abnormal changes of inflammatory cells and inflammatory cytokines (IL-1ß, IL-6, TGF-ß1, TNF-α, IFN-γ, IL-10, CXCL-1 and CXCL-2) in bronchoalveolar lavage fluid (BALF), pulmonary tissue and cell culture supernatant. Furthermore, NiO NPs activated the p38 MAPK pathway and downregulated MEG3 in vivo and in vitro. However, p38 MAPK pathway inhibitor (10 µM SB203580) reversed the alterations in the expression levels of inflammatory cytokines induced by NiO NPs. Meanwhile, over-expressed MEG3 significantly suppressed NiO NPs-induced p38 MAPK pathway activation and inflammatory cytokines changes. Overall, the above results proved that over-expression of lncRNA MEG3 reduced NiO NPs-induced inflammatory damage by preventing the activation of p38 MAPK pathway.


Assuntos
Nanopartículas , RNA Longo não Codificante , Animais , Pulmão/metabolismo , RNA Longo não Codificante/genética , Ratos , Ratos Wistar , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
Environ Toxicol ; 37(1): 79-91, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34608745

RESUMO

Long noncoding RNA maternally expressed gene 3 (lncRNA MEG3) was down-regulated in pulmonary fibrosis of rats induced by Nickel oxide nanoparticles (NiO NPs), while the downstream regulatory mechanisms of MEG3 remain unclear. This study aimed to investigate the relationship among MEG3, Hedgehog (Hh) signaling pathway and autophagy in pulmonary fibrosis caused by NiO NPs. The pulmonary fibrosis model in rats was constructed by intratracheal instillation of 0.015, 0.06, and 0.24 mg/kg NiO NPs twice a week for 9 weeks. Collagen deposition model was established by treating A549 cells with 25, 50, and 100 µg/mL NiO NPs for 24 h. Our results indicated that NiO NPs activated Hh pathway, down-regulated the expression of MEG3, and reduced autophagy activity in vivo and in vitro. Meanwhile, the autophagy process was promoted by Hh pathway inhibitor (CDG-0449), while the collagen formation in A549 cells was reduced by autophagy activator (Rapamycin). Furthermore, the overexpressed MEG3 inhibited the activation of Hh pathway, resulting in autophagy activity enhancement along with collagen formation reduction. In summary, lncRNA MEG3 can restrain pulmonary fibrosis induced by NiO NPs via regulating hedgehog signaling pathway-mediated autophagy, which may serve as a potential therapeutic strategy for pulmonary fibrosis.


Assuntos
Fibrose Pulmonar , RNA Longo não Codificante , Animais , Autofagia , Proteínas Hedgehog/genética , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/genética , RNA Longo não Codificante/genética , Ratos , Transdução de Sinais
9.
Environ Toxicol ; 36(6): 1099-1110, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33547861

RESUMO

Nickel oxide nanoparticles (NiO NPs) causes pulmonary fibrosis via activating transforming growth factor-ß1 (TGF-ß1) in rats, but its upstream regulatory mechanisms are unknown. This study aimed to explore the role of long noncoding RNA (lncRNA) maternally expressed gene 3 (MEG3) in NiO NPs-induced collagen deposition. Male Wistar rats were intratracheally instilled with NiO NPs (0.015, 0.06, and 0.24 mg/kg b.w.) twice a week for 9 weeks. Human lung adenocarcinoma epithelial cells (A549 cells) were cultured with NiO NPs (25, 50, and 100 µg/ml) to establish collagen deposition model. We discovered that NiO NPs-induced rat pulmonary fibrosis was accompanied by the epithelial-mesenchymal transition (EMT) occurrence and MEG3 down-regulation in rat lung tissues. In cell collagen deposition model, NiO NPs also evoked EMT and decreased MEG3 expression in a dose-dependent manner in A549 cells. By overexpressing MEG3 in A549 cells, we found that MEG3 inhibited the level of TGF-ß1, EMT process and collagen formation. Moreover, our data showed that SB431542 (TGF-ß1 inhibitor) had an inhibitory effect on NiO NPs-induced EMT and collagen formation. Our results indicated that MEG3 inhibited NiO NPs-induced collagen deposition by regulating TGF-ß1-mediated EMT process, which may provide some clues for insighting into the mechanisms of NiO NPs-induced pulmonary fibrosis.


Assuntos
Nanopartículas , Fibrose Pulmonar , RNA Longo não Codificante , Animais , Transição Epitelial-Mesenquimal , Masculino , Níquel , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/genética , RNA Longo não Codificante/genética , Ratos , Ratos Wistar , Fator de Crescimento Transformador beta1/genética
10.
Environ Toxicol ; 35(12): 1308-1317, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32681694

RESUMO

Our previous study demonstrated that nano nickel oxide (NiO) induce pulmonary fibrosis in rats and collagen excessive formation in A549 cells, which mechanism was related with the increasing transforming growth factor ß1 (TGF-ß1) secretion. However, it remains unclear understanding the role of TGF-ß1 in collagen excessive formation. Here, we found nano NiO could directly promote epithelial-mesenchymal transition (EMT) via the TGF-ß1/Smads pathway in A549 cells. First, cytotoxicity induced by nano NiO has a dose- and time-dependent manner according to methylthiaozol tetrazolium assay. Second, nano NiO led to the increased contents of type I collagen (Col-I), TGF-ß1, p-Smad2, p-Smad3, alpha-smooth muscle actin (α-SMA), vimentin, and fibronectin, indicating Smads pathway activation and EMT occurence. Third, to verify whether TGF-ß1 activated Smads signaling pathway and EMT occurence, A549 cells were exposed to nano NiO and TGF-ß1 inhibitors (10 µM SB431542). The results showed that TGF-ß1 inhibitors alleviated the nano NiO-induced cytotoxicity and Col-I excessive formation. Meanwhile, TGF-ß1 inhibitors reversed the proteins expression trends of Col-I, p-Smad2, p-Smad3, α-SMA, vimentin, fibronectin, and E-cadherin. These observations suggested that EMT occurrence via TGF-ß1/Smads pathway might play an important role in the collagen excessive formation induced by nano NiO in A549 cells.


Assuntos
Transição Epitelial-Mesenquimal/efeitos dos fármacos , Nanopartículas/toxicidade , Níquel/toxicidade , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Células A549 , Técnicas de Cultura de Células , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Transdução de Sinais
11.
Environ Toxicol ; 35(4): 419-429, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31737983

RESUMO

Nickel oxide nanoparticles (Nano NiO) bears hepatotoxicity, while whether it leads to liver fibrosis remains unclear. The aim of this study was to establish the Nano NiO-induced hepatic fibrosis model in vivo and investigate the roles of transforming growth factor ß1 (TGF-ß1) in Smad pathway activation, epithelial-mesenchymal transition (EMT) occurrence, and extracellular matrix (ECM) deposition in vitro. Male Wistar rats were exposed to 0.015, 0.06, and 0.24 mg/kg Nano NiO by intratracheal instilling twice a week for 9 weeks. HepG2 cells were treated with 100 µg/mL Nano NiO and TGF-ß1 inhibitor (SB431542) to explore the mechanism of collagen formation. Results of Masson staining as well as the elevated levels of type I collagen (Col-I) and Col-III suggested that Nano NiO resulted in hepatic fibrosis in rats. Furthermore, Nano NiO increased the protein expression of TGF-ß1, p-Smad2, p-Smad3, alpha-smooth muscle actin (α-SMA), matrix metalloproteinase9 (MMP9), and tissue inhibitors of metalloproteinase1 (TIMP1), while decreased the protein content of E-cadherin and Smad7 in rat liver and HepG2 cells. Most importantly, Nano NiO-triggered the abnormal expression of the abovementioned proteins were all alleviated by co-treatment with SB431542, implying that TGF-ß1-mediated Smad pathway, EMT and MMP9/TIMP1 imbalance were involved in overproduction of collagen in HepG2 cells. In conclusion, these findings indicated that Nano NiO induced hepatic fibrosis via TGF-ß1-mediated Smad pathway activation, EMT occurrence, and ECM deposition.


Assuntos
Transição Epitelial-Mesenquimal/efeitos dos fármacos , Cirrose Hepática/induzido quimicamente , Nanopartículas/química , Níquel/toxicidade , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Actinas/metabolismo , Animais , Colágeno Tipo I/metabolismo , Células Hep G2 , Humanos , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Masculino , Níquel/química , Ratos , Ratos Wistar , Transdução de Sinais
12.
Environ Toxicol ; 34(6): 719-727, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30810263

RESUMO

Nickel oxide nanoparticles (Nano NiO) could induce pulmonary fibrosis, however, the mechanisms are still unknown. The aim of the present study was to explore the roles of transforming growth factor-ß1 (TGF-ß1), mitogen-activated protein kinase (MAPK) pathway and MMPs/TIMPs balance in Nano NiO-induced pulmonary fibrosis. For that purpose, we first established Nano NiO-induced human lung adenocarcinoma epithelial cells (A549 cells) model of collagen excessive formation through detecting the levels of hydroxyproline (Hyp) and type I collagen (Col-I). Then the protein levels of TGF-ß1, MAPKs, and MMPs/TIMPs were assessed by Western blot. The results showed that Nano NiO resulted in the increased contents of Hyp, Col-I, and TGF-ß1, the MAPK pathway activation and MMPs/TIMPs imbalance with a dose-dependent manner. In addition, to investigate whether TGF-ß1 mediated MAPK signaling pathway, A549 cells were treated by 100 µg/mL Nano NiO combined with TGF-ß1, p38 MAPK, and ERK1/2 inhibitors (10 µM SB431542, 10 µM SB203580, and 10 µM U0126), respectively. We found that MAPK signal pathway was suppressed by TGF-ß1 inhibitor. Meanwhile, the increased contents of Hyp and Col-I, and MMPs/TIMPs imbalance were alleviated by the p38 MAPK and ERK1/2 inhibitors, respectively. These findings indicated that the MAPK pathway and MMPs/TIMPs imbalance were involved in collagen excessive formation induced by Nano NiO.


Assuntos
Colágeno Tipo I/biossíntese , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Nanopartículas/toxicidade , Níquel/toxicidade , Fator de Crescimento Transformador beta1/metabolismo , Células A549 , Humanos , Hidroxiprolina/biossíntese , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fibrose Pulmonar/induzido quimicamente
13.
Environ Toxicol ; 32(12): 2492-2499, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28945320

RESUMO

Nickel oxide nanoparticles (nano NiO) could induce hepatocyte apoptosis, while its potential mechanisms are unclear. This study aimed to explore the role of endoplasmic reticulum (ER) stress pathways in hepatocyte apoptosis induced by nano NiO. Male Wistar rats were administrated with nano NiO (0.015, 0.06, and 0.24 mg/kg b.w.) and micro NiO (0.24 mg/kg b.w.) by intratracheal instillation twice a week for 6 weeks. We measured the hepatocyte apoptosis levels by TdT-mediated dUTP nick-end labeling (TUNEL) staining, ER stress related gene and protein expression levels in rat liver. The results showed that the TUNEL positive cells increased after exposure nano NiO, hinting hepatocyte apoptosis. The up-regulated gene and protein levels of 78 kD glucose regulated protein and CCAAT/enhancer binding protein homologous protein suggested that nano NiO triggered ER stress. Nano NiO exposure contributed to the increased protein contents of inositol-requiring enzyme 1 (IRE-1)α, p-IRE-1α, X box protein-1S, pancreatic ER kinase (PERK), p-PERK, eukaryotic initiation factor-2 alpha (eIF-2α), p-eIF-2α, caspase-12, -9, and -3, implicating that nano NiO can activate the pathways of ER stress-mediated apoptosis. These findings indicate that the ER stress pathways may play an important role in hepatocyte apoptosis induced by nano NiO.


Assuntos
Apoptose/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Nanopartículas Metálicas/toxicidade , Níquel/toxicidade , Animais , Caspases/metabolismo , Endorribonucleases/metabolismo , Fator de Iniciação 2 em Eucariotos/metabolismo , Hepatócitos/metabolismo , Masculino , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Ratos Wistar , eIF-2 Quinase/metabolismo
14.
Environ Toxicol ; 32(4): 1354-1362, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27464988

RESUMO

With the progress of nanotechnology, nano nickel oxide (NiO) has been extensively used as sensors, battery electrodes, catalysts, and cosmetics. Previous researches verified that nano NiO could exert pulmonary toxicity, but its mechanism was unclear. To shed light upon this, the role of nuclear factor-κB (NF-κB) activation and Th1/Th2 imbalance were to explore in pulmonary damage induced by nano NiO. Male Wistar rats were randomized into control group, nano NiO groups (0.015, 0.06, and 0.24 mg kg-1 ) and micro NiO group (0.024 mg kg-1 ) and treated by intratracheal instillation twice a week for 6 weeks. The results showed that the abnormal changes induced by nano NiO were found on indicators of nitrative stress (NO, TNOS, and iNOS), inflammatory cytokines (TNF-α, IL-2, and IL-10) and cytokine-induced neutrophil chemoattractants (CINC-1, CINC-2αß, and CINC-3) in lung tissue. In addition, nano NiO instillation induced the upregulated mRNA and protein expression of NF-κB, inhibitor of κB kinase-α (IKK-α) and nuclear factor-inducing kinase (NIK). The protein content of GATA-3 increased as well as T-bet decreased in nano NiO groups, and the ratio of T-bet/GATA-3, as a key evaluation indicator of Th1/Th2 balance, was lower than the control group. The findings indicated that nano NiO could enhance the nitrative stress and inflammatory response in lung tissue, and its mechanism was related to the NF-κB activation and Th1/Th2 imbalance. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 1354-1362, 2017.


Assuntos
Poluentes Ambientais/toxicidade , Pulmão/efeitos dos fármacos , Nanopartículas Metálicas/toxicidade , NF-kappa B/metabolismo , Níquel/toxicidade , Animais , Citocinas/metabolismo , Fator de Transcrição GATA3/genética , Fator de Transcrição GATA3/metabolismo , Pulmão/metabolismo , Pulmão/patologia , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/imunologia , Lesão Pulmonar/metabolismo , Masculino , Ratos , Ratos Wistar , Células Th1/efeitos dos fármacos , Células Th2/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
15.
Environ Toxicol ; 32(7): 1918-1926, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28296042

RESUMO

Nickel can induce apoptosis of testicular Leydig cells in mice, whereas the mechanisms remain unclear. In this study, we investigated the role of nickel-induced reactive oxygen species (ROS) generation in mitochondria and endoplasmic reticulum stress (ERS) mediated apoptosis pathways in rat Leydig cells. Fluorescent DCF and Annexin-V FITC/PI staining were performed to measure the production of ROS and apoptosis in Leydig cells. RT-qPCR and Western blot were conducted to analyze the key genes and proteins involved in mitochondria and ERS apoptotic pathways. The results showed that nickel sulfate induced ROS generation, consequently resulted in nucleolus deformation and apoptosis in testicular Leydig cells, which were then attenuated by ROS inhibitors of N-acetylcysteine (NAC) and 2,2,6,6-tetramethyl-1-piperidinyloxy (TEMPO). Nickel sulfate-triggered Leydig cells apoptosis via mitochondria and ERS pathways was characterized by the upregulated mRNA and proteins expression of Bak, cytochrome c, caspase 9, caspase 3, GRP78, GADD153, and caspase 12, which were inhibited by NAC and TEMPO respectively. The findings indicated that nickel-induced ROS generation was involved in apoptosis via mitochondria and ERS pathways in rat Leydig cells.


Assuntos
Estresse do Retículo Endoplasmático/efeitos dos fármacos , Células Intersticiais do Testículo/efeitos dos fármacos , Mitocôndrias/metabolismo , Níquel/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Acetilcisteína/farmacologia , Animais , Apoptose/efeitos dos fármacos , Caspase 12/metabolismo , Caspase 3/metabolismo , Caspase 9/metabolismo , Óxidos N-Cíclicos/farmacologia , Citocromos c/metabolismo , Chaperona BiP do Retículo Endoplasmático , Proteínas de Choque Térmico/metabolismo , Células Intersticiais do Testículo/metabolismo , Masculino , Ratos Wistar , Espécies Reativas de Oxigênio/antagonistas & inibidores , Transdução de Sinais , Fator de Transcrição CHOP/metabolismo
16.
Wei Sheng Yan Jiu ; 46(5): 797-801, 2017 Sep.
Artigo em Zh | MEDLINE | ID: mdl-29903311

RESUMO

OBJECTIVE: To estimate the metabolic parameters in different tissues and organs, build the physiologically based pharmacokinetic( PBPK) model of rat and occupational population, and predict the toxic dynamic characteristics exposure to nickel. METHODS: The partition coefficients in different tissues and organs were estimated using vector datas of nickel by the optimization and statistics files of acslx software. The PBTK model of occupational population exposure to nickel was built according to the metabolic parameters by acslx software. RESULTS: The evaluated partition coefficient of nickel were kidney blood( 0. 668), lung blood( 0. 102), spleen blood( 0. 037), liver blood( 0. 028), heart blood( 0. 022), and brain blood( 0. 006). The constructed successful PBPK model of occupational population exposed to 0. 1 mg/m~3 nickel for 8 hours showed that the nickel concentration is higher in kidney reached at 3. 328 µg/kg, followed by the spleen( 0. 185 µg/kg), liver( 0. 140 µg/kg) and heart( 0. 110 µg/kg). The content of nickel is lower in the brain( 0. 030 µg/kg). The kidneys is the major metabolic organs for nickel. CONCLUSION: The PBPK model can be used to convert the nickel levels from external exposure to internal exposure for each organ and to evaluate the time-dose relationship exposure to nickel in both rat and occupational population studies.


Assuntos
Modelos Biológicos , Níquel/farmacocinética , Níquel/toxicidade , Exposição Ocupacional , Toxicocinética , Animais , Ratos , Software
17.
Wei Sheng Yan Jiu ; 45(4): 563-567, 2016 Jul.
Artigo em Zh | MEDLINE | ID: mdl-29903323

RESUMO

OBJECTIVE: To investigate the subchronic lung injury induced by nano nickel oxide( nano NiO) and its mechanism from the view of nitrative stress in rats. METHODS: A total of 40 adult male Wistar rats were randomly divided into 5 groups, control group( normal saline), 0. 015, 0. 06 and 0. 24 mg / kg nano NiO groups and 0. 24 mg / kg micro NiO group. Rats received intratracheally instilled nano NiO, micro NiO and normal saline twice a week for 6 weeks, respectively. All rats were sacrificed after the exposure to obtain lung tissues. HE staining was used to observe the lung pathological changes. The content of nitric oxide, and the activities of total nitric oxide synthase( TNOS) and inducible nitric oxide synthase( iNOS) in pulmonary tissue homogenate were measured by microplate reader. The levels of interleukin-2( IL-2), transforminggrowth factor-beta( TGF-ß), interferon-gamma( IFN-γ) and 8-hydroxy-2'-deoxyguanosine( 8-OHd G) in serum were detected by enzyme-linked immunosorbent assay( ELISA). RESULTS: The results of lung histopathology showed that the widened alveolar speta, inflammatory infiltration and nanoparticles deposition increased with the increasing dosage of nano NiO. Compared to control group, the content of NO and the activities of TNOS and iNOS in 0. 24 mg / kg nano NiO group increased in lung homogenate( P < 0. 05). The levels of IL-2, TGF-ß and IFN-γ in nano NiO 0. 06 and 0. 24 mg /kg group were higher than that of control group, and the level of 8-OHd G increased in nano NiO 0. 24 mg / kg group when compared to control group in serum( P < 0. 05). Compared to micro NiO group, the levels of NO and iNOS in lung homogenate, and the serum levels of IL-2 and 8-OHd G increased after exposed to 0. 24 mg / kg nano NiO in rats( P < 0. 05). CONCLUSION: Nano NiO can lead to lung injury in rats which may be related with nitrative stress reaction based on pulmonary inflammation.


Assuntos
Interleucina-2/sangue , Lesão Pulmonar/induzido quimicamente , NF-kappa B/metabolismo , Níquel/toxicidade , Óxido Nítrico Sintase Tipo II , Animais , Pulmão , Masculino , Óxido Nítrico , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Fator de Necrose Tumoral alfa/metabolismo
18.
J Nanosci Nanotechnol ; 15(2): 1135-42, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26353623

RESUMO

In an effort to examine liver, heart and kidney injury, immune response, and other physiological effect in rats caused by intratracheal instillation of nano titanium dioxide (TiO2) for 28 days, we assessed T lymphocytes counts, hematological indices, biochemical parameters, cytokines assay and histopathological changes in nano TiO2 treated rats. Indeed, rats treated with nano TiO2 displayed a reduction in body weight and coefficients of the hearts. Edema and loose cytoplasm on liver cells were found in nano groups. The results showed that a statistically significant increased in the BUN, HTC and AST levels than those in control group. Our data suggested that the immunologically competent cells of CD3+, CD4+, and CD8+ caused by nano TiO2 32 mg/kg group were significantly lower than control group. The ratio of CD4+ to CD8+ from the nano 32 mg/kg group was significantly increased and showed a disturbance of cellular immune function. But ELISA analysis showed that no significant changes in IFN-γ and IL-4 were observed throughout the experimental period in this study.


Assuntos
Citocinas/imunologia , Nanopartículas Metálicas/toxicidade , Nanopartículas Metálicas/ultraestrutura , Síndrome de Resposta Inflamatória Sistêmica/induzido quimicamente , Síndrome de Resposta Inflamatória Sistêmica/imunologia , Titânio/toxicidade , Animais , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/imunologia , Teste de Materiais , Nanopartículas Metálicas/química , Tamanho da Partícula , Ratos , Titânio/química
19.
Int J Mol Sci ; 15(4): 6961-73, 2014 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-24758935

RESUMO

The potential immune effects of titanium dioxide nanoparticles (nano-TiO2) are raising concern. Our previous study verified that nano-TiO2 induce local immune response in lung tissue followed by intratracheal instillation administration. In this study, we aim to evaluate the systemic immune effects of nano-TiO2. Sprague Dawley rats were treated by intratracheal instillation with nano-TiO2 at doses of 0.5, 4, and 32 mg/kg body weight, micro-TiO2 with 32 mg/kg body weight and 0.9% NaCl, respectively. The exposure was conducted twice a week, for four consecutive weeks. Histopathological immune organs from exposed animals showed slight congestion in spleen, generally brown particulate deposition in cervical and axillary lymph node. Furthermore, immune function response was characterized by increased proliferation of T cells and B cells following mitogen stimulation and enhanced natural killer (NK) cell killing activity in spleen, accompanying by increased number of B cells in blood. No significant changes of Th1-type cytokines (IL-2 and INF-γ) and Th2-type cytokines (TNF-α and IL-6) were observed. Intratracheal exposure to nano-TiO2 may be one of triggers to be responsible for the systemic immune response. Further study is needed to confirm long-lasting lymphocyte responses and the potential mechanisms.


Assuntos
Nanopartículas/química , Titânio/química , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Proliferação de Células/efeitos dos fármacos , Instilação de Medicamentos , Interleucina-6/sangue , Células Matadoras Naturais/citologia , Células Matadoras Naturais/imunologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Linfonodos/patologia , Masculino , Nanopartículas/toxicidade , Ratos , Ratos Sprague-Dawley , Baço/citologia , Baço/patologia , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Fator de Necrose Tumoral alfa/sangue
20.
Biomed Pharmacother ; 175: 116698, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38713946

RESUMO

Neurotoxicity can cause a range of symptoms and disorders in humans, including neurodegenerative diseases, neurodevelopmental disorders, nerve conduction abnormalities, neuroinflammation, autoimmune disorders, and cognitive deficits. The cyclic guanosine-adenosine synthase (cGAS)-stimulator of interferon genes (STING) pathway and NF-κB pathway are two important signaling pathways involved in the innate immune response. The cGAS-STING pathway is activated by the recognition of intracellular DNA, which triggers the production of type I interferons and pro-inflammatory cytokines, such as tumor necrosis factor, IL-1ß, and IL-6. These cytokines play a role in oxidative stress and mitochondrial dysfunction in neurons. The NF-κB pathway is activated by various stimuli, such as bacterial lipopolysaccharide, viral particle components, and neurotoxins. NF-κB activation may lead to the production of pro-inflammatory cytokines, which promote neuroinflammation and cause neuronal damage. A potential interaction exists between the cGAS-STING and NF-κB pathways, and NF-κB activation blocks STING degradation by inhibiting microtubule-mediated STING transport. This review examines the progress of research on the roles of these pathways in neurotoxicity and their interrelationships. Understanding the mechanisms of these pathways will provide valuable therapeutic insights for preventing and controlling neurotoxicity.


Assuntos
Proteínas de Membrana , NF-kappa B , Nucleotidiltransferases , Transdução de Sinais , Humanos , NF-kappa B/metabolismo , Nucleotidiltransferases/metabolismo , Proteínas de Membrana/metabolismo , Animais , Síndromes Neurotóxicas/metabolismo , Síndromes Neurotóxicas/etiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA