Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Arch Toxicol ; 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38806717

RESUMO

A mechanism exploration is an important part of toxicological studies. However, traditional cell and animal models can no longer meet the current needs for in-depth studies of toxicological mechanisms. The three-dimensional (3D) organoid derived from human embryonic stem cells (hESC) or induced pluripotent stem cells (hiPSC) is an ideal experimental model for the study of toxicological effects and mechanisms, which further recapitulates the human tissue microenvironment and provides a reliable method for studying complex cell-cell interactions. This article provides a comprehensive overview of the state of the 3D organoid technology in toxicological studies, including a bibliometric analysis of the existing literature and an exploration of the latest advances in toxicological mechanisms. The use of 3D organoids in toxicology research is growing rapidly, with applications in disease modeling, organ-on-chips, and drug toxicity screening being emphasized, but academic communications among countries/regions, institutions, and research scholars need to be further strengthened. Attempts to study the toxicological mechanisms of exogenous chemicals such as heavy metals, nanoparticles, drugs and organic pollutants are also increasing. It can be expected that 3D organoids can be better applied to the safety evaluation of exogenous chemicals by establishing a standardized methodology.

2.
J Appl Toxicol ; 2024 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-38642013

RESUMO

Mitophagy is a selective cellular process critical for the removal of damaged mitochondria. It is essential in regulating mitochondrial number, ensuring mitochondrial functionality, and maintaining cellular equilibrium, ultimately influencing cell destiny. Numerous pathologies, such as neurodegenerative diseases, cardiovascular disorders, cancers, and various other conditions, are associated with mitochondrial dysfunctions. Thus, a detailed exploration of the regulatory mechanisms of mitophagy is pivotal for enhancing our understanding and for the discovery of novel preventive and therapeutic options for these diseases. Nanomaterials have become integral in biomedicine and various other sectors, offering advanced solutions for medical uses including biological imaging, drug delivery, and disease diagnostics and therapy. Mitophagy is vital in managing the cellular effects elicited by nanomaterials. This review provides a comprehensive analysis of the molecular mechanisms underpinning mitophagy, underscoring its significant influence on the biological responses of cells to nanomaterials. Nanoparticles can initiate mitophagy via various pathways, among which the PINK1-Parkin pathway is critical for cellular defense against nanomaterial-induced damage by promoting mitophagy. The role of mitophagy in biological effects was induced by nanomaterials, which are associated with alterations in Ca2+ levels, the production of reactive oxygen species, endoplasmic reticulum stress, and lysosomal damage.

3.
J Appl Toxicol ; 43(1): 167-185, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-35995895

RESUMO

Airborne particulate matter (PM), the primary component associated with health risks in air pollution, can negatively impact human health. Studies have shown that PM can enter the brain by inhalation, but data on the exact quantity of particles that reach the brain are unknown. Particulate matter exposure can result in neurotoxicity. Exposure to PM poses a greater health risk to infants and children because their nervous systems are not fully developed. This review paper highlights the association between PM and neurodevelopmental toxicity (NDT). Exposure to PM can induce oxidative stress and inflammation, potentially resulting in blood-brain barrier damage and increased susceptibility to development of neurodevelopmental disorders (NDD), such as autism spectrum disorders and attention deficit disorders. In addition, human and animal exposure to PM can induce microglia activation and epigenetic alterations and alter the neurotransmitter levels, which may increase risks for development of NDD. However, the systematic comparisons of the effects of PM on NDD at different ages of exposure are deficient. The elucidation of PM exposure risks and NDT in children during the early developmental stages are of great importance. The synthesis of current research may help to identify markers and mechanisms of PM-induced neurodevelopmental toxicity, allowing for the development of strategies to prevent permanent damage of developing brain.


Assuntos
Poluentes Atmosféricos , Poluição do Ar , Síndromes Neurotóxicas , Animais , Criança , Humanos , Material Particulado/toxicidade , Poluentes Atmosféricos/toxicidade , Poluição do Ar/efeitos adversos , Estresse Oxidativo , Síndromes Neurotóxicas/etiologia
4.
J Appl Toxicol ; 43(8): 1242-1252, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-36918407

RESUMO

Recombinant human metallothionein III (rh-MT-III) is a genetically engineered product produced by Escherichia coli fermentation technology. Its molecules contain abundant reducing sulfhydryl groups, which possess the ability to bind heavy metal ions. The present study was to evaluate the binding effects of rh-MT-III against copper and cadmium in vitro and to investigate the antioxidant activity of rh-MT-III using Caenorhabditis elegans in vivo. For in vitro experiments, the binding rates of copper and cadmium were 91.4% and 97.3% for rh-MT-III at a dosage of 200 µg/mL at 10 h, respectively. For in vivo assays, the oxidative stress induced by copper (CuSO4 , 10 µg/mL) and cadmium (CdCl2 , 10 µg/mL) was significantly reduced after 72 h of exposure to different doses of rh-MT-III (5-500 µg/mL), indicated by restoring locomotion behavior and growth, and reducing malondialdehyde and reactive oxygen species levels in C. elegans. Moreover, rh-MT-III decreased the deposition of lipofuscin and fat content, which could delay the progression of aging. In addition, rh-MT-III (500 µg/mL) promoted the up-regulation of Mtl-1 and Mtl-2 gene expression in C. elegans, which could enhance the resistance to oxidative stress by increasing the enzymatic activity of antioxidant defense system and scavenging free radicals. The results indicated that supplemental rh-MT-III could effectively protect C. elegans from heavy metal stress, providing an experimental basis for the future application and development of rh-MT-III.


Assuntos
Cádmio , Metais Pesados , Animais , Humanos , Cádmio/toxicidade , Cádmio/metabolismo , Cobre , Metalotioneína 3 , Caenorhabditis elegans , Metalotioneína/genética , Metalotioneína/metabolismo , Estresse Oxidativo , Antioxidantes/farmacologia , Antioxidantes/metabolismo
5.
J Appl Toxicol ; 42(5): 738-749, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34708887

RESUMO

Fine particulate matter (PM2.5 )-induced detrimental cardiovascular effects have been widely concerned, especially for endothelial cells, which is the first barrier of the cardiovascular system. Among potential mechanisms involved, reactive oxidative species take up a crucial part. However, source of oxidative stress and its relationship with inflammatory response have been rarely studied in PM2.5 -induced endothelial injury. Here, as a key oxidase that catalyzes redox reactions, NADPH oxidase (NOX) was investigated. Human umbilical vein endothelial cells (EA.hy926) were exposed to Standard Reference Material 1648a of urban PM2.5 for 24 h, which resulted in NOX-sourced oxidative stress, endothelial dysfunction, and inflammation induction. These are manifested by the up-regulation of NOX, increase of superoxide anion and hydrogen peroxide, elevated endothelin-1 (ET-1) and asymmetric dimethylarginine (ADMA) level, reduced nitric oxide (NO) production, and down-regulation of phosphorylation of endothelial NO synthase (eNOS) with increased levels of inducible NO synthase, as well as the imbalance between tissue-type plasminogen activator (tPA) and plasminogen activator inhibitor 1 (PAI-1), and changes in the levels of pro-inflammatory and anti-inflammatory factors. However, administration of NOX1/4 inhibitor GKT137831 alleviated PM2.5 -induced elevated endothelial dysfunction biomarkers (NO, ET-1, ADMA, iNOS, and tPA/PAI-1), inflammatory factors (IL-1ß, IL-10, and IL-18), and adhesion molecules (ICAM-1, VCAM-1, and P-selectin) and also passivated NOX-dependent AKT and eNOS phosphorylation that involved in endothelial activation. In summary, PM2.5 -induced NOX up-regulation is the source of ROS in EA.hy926, which activated AKT/eNOS/NO signal response leading to endothelial dysfunction and inflammatory damage in EA.hy926 cells.


Assuntos
NADPH Oxidases , Óxido Nítrico , Células Endoteliais da Veia Umbilical Humana , Humanos , Material Particulado/toxicidade , Inibidor 1 de Ativador de Plasminogênio/farmacologia , Proteínas Proto-Oncogênicas c-akt , Espécies Reativas de Oxigênio
6.
J Appl Toxicol ; 41(1): 65-81, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32686875

RESUMO

With the development of nanotechnology, metal-containing nanoparticles are used widely in the diagnosis, monitoring and treatment of central nervous system (CNS) diseases. The neurotoxicity of these nanoparticles has drawn attention. Glial cells (particularly microglial cells and astrocytes) have important functions in the CNS. Neural disorders are related to functional/histologic damage to glial cells. Dysfunctions of microglial cells or astrocytes injure the brain, and cause the neurodegeneration seen in Alzheimer's disease and Parkinson's disease. We have summarized the route of access of metal-containing nanoparticles to the CNS, as well as their neurotoxicity and potential molecular mechanisms involved in glial cells. Metal-containing nanoparticles cross or bypass the blood-brain barrier, access the CNS and cause neurotoxicity. The potential mechanisms are related to inflammation, oxidative stress, DNA and/or mitochondrial damage and cell death, all of which are mediated by microglial cell activation, inflammatory factor release, generation of reactive oxygen species, apoptosis and/or autophagy in glial cells. Moreover, these processes increase the burden of the CNS and even accelerate the occurrence or development of neurodegenerative diseases. Some important signaling pathways involved in the mechanism of neurotoxicity in glial cells caused by nanoparticles are also discussed.


Assuntos
Morte Celular/efeitos dos fármacos , Doenças do Sistema Nervoso Central/induzido quimicamente , Doenças do Sistema Nervoso Central/terapia , Nanopartículas Metálicas/toxicidade , Nanopartículas Metálicas/uso terapêutico , Neuroglia/efeitos dos fármacos , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/fisiopatologia , Humanos
7.
J Appl Toxicol ; 41(12): 2055-2067, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-33993517

RESUMO

Silver nanoparticles (AgNPs) have become widespread in the environment with increasing industrial applications. But the studies about their potential health risks are far from enough, especially in neurotoxic effects. This study aimed to investigate the neurotoxic effects of longer-term exposure (prolonged exposure for 48 h and chronic exposure for 6 days) of 20nm AgNPs with/without polyvinylpyrrolidone (PVP) coating at low concentrations (0.01-10 mg·L-1 ) to Caenorhabditis elegans. The results suggested that exposure to AgNPs induced damage to nematode survival, with the longest and relative average life span reduced. Exposure to AgNPs caused neurotoxicity on locomotion behaviors (head thrashes, body bends, pharyngeal pumping frequency, and defecation interval) and sensory perception behaviors (chemotaxis assay and thermotaxis assay), as well as impaired dopaminergic, GABAergic, and cholinergic neurons, except for glutamatergic, based on the alters fluorescence intensity, in a dose- and time-dependent manner. Further investigations suggested that the low-dose AgNPs (0.01-0.1 mg·L-1 ) exposure raises receptors of GABAergic and dopamine in C. elegans at the genetic level, whereas opposite results were observed at higher doses (1-10 mg·L-1 ), which implied that AgNPs could cause neurotoxicity by impairing neurotransmitter delivery. The PVP-AgNPs could cause a higher fatality rate and neurotoxicity at the same dose. Notably, AgNPs did not cause any deleterious effect on nematodes at the lowest dose of 0.01 mg·L-1 . In general, these results suggested that AgNPs possess the neurotoxic potential in C. elegans and provided useful information to understand the neurotoxicity of AgNPs, which would offer an inspiring perspective on the safe application.


Assuntos
Caenorhabditis elegans/efeitos dos fármacos , Nanopartículas Metálicas/toxicidade , Neurônios/efeitos dos fármacos , Povidona/toxicidade , Prata/toxicidade , Animais , Caenorhabditis elegans/fisiologia , Neurônios/fisiologia , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/fisiopatologia , Excipientes Farmacêuticos/toxicidade , Substitutos do Plasma/toxicidade
8.
Ecotoxicol Environ Saf ; 221: 112452, 2021 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-34198186

RESUMO

Helicoverpa armigera (cotton bollworm) is one of the most destructive pests worldwide. Due to resistance to Bacillus thuringiensis and conventional insecticides, an effective management strategy to control this pest is urgently needed. Spinosad, a natural pesticide, is considered an alternative; however, the mechanism underlying the developmental effects of sublethal spinosad exposure remains elusive. In this study, the mechanism was examined using an insect model of H. armigera. Results confirmed that exposure to sublethal spinosad led to reduced larval wet weight, delayed larval developmental period, caused difficulty in molting, and deformed pupae. Further investigation demonstrated that exposure to sublethal spinosad caused a significant decrease in 20E titer and increase in JH titer, thereby leading to the discordance between 20E and JH titers, and consequently alteration in the expression levels of HR3 and Kr-h1. These results suggested that sublethal spinosad caused hormonal disorders in larvae, which directly affect insect development. Our study serves as a reference and basis for the toxicity evaluation of spinosad on molting and pupation in insect metamorphosis, which may contribute to identifying targets for effective control of cotton bollworm.


Assuntos
Inseticidas/toxicidade , Macrolídeos/toxicidade , Mariposas/efeitos dos fármacos , Animais , Combinação de Medicamentos , Larva/efeitos dos fármacos , Muda/efeitos dos fármacos , Mariposas/crescimento & desenvolvimento , Pupa/efeitos dos fármacos
9.
Ecotoxicol Environ Saf ; 208: 111463, 2021 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-33130480

RESUMO

With the increasing use of silver nanoparticles (AgNPs) in biological materials, the cytotoxicity caused by these particles has attracted much attention. However, the molecular mechanism underlying AgNP cytotoxicity remains unclear. In this study, we aimed to systematically investigate the toxicity induced by AgNP exposure to the lung adenocarcinoma A549 cell line at the subcellular and signaling pathway levels and elucidate the related molecular mechanism. The survival rate of cells exposed to AgNPs at 0, 20, 40, 80, and 160 µg/mL for 24 or 48 h decreased in a dose- and time-dependent manner. AgNPs induced autophagy and mitophagy, determined by the transmission electron microscopy investigation and upregulation of LC3 II/I, p62, PINK1, and Parkin expression levels. AgNP treatment induced lysosomal injury, including the decline of lysosomal membrane integrity and increase in cathepsin B level. The decreased in mitochondrial membrane potential, along with upregulation of cytochrome c, caspases 9 and 3, and BAX/BCL2, further suggested that mitochondrial injury were involved in AgNP-induced apoptosis. In addition, mitochondrial injury may further lead to excessive production of reactive oxygen species and oxidative/ antioxidant imbalance. The results suggested that AgNPs could regulate autophagy via mitochondrial and lysosome injury in A549 cells. The information of the molecular mechanism will provide an experimental basis for the safe application of nanomaterials.


Assuntos
Nanopartículas Metálicas/toxicidade , Mitofagia/fisiologia , Prata/toxicidade , Células A549 , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Caspase 9 , Morte Celular/efeitos dos fármacos , Humanos , Lisossomos/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitofagia/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Ubiquitina-Proteína Ligases
10.
Ecotoxicol Environ Saf ; 208: 111696, 2021 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-33396027

RESUMO

With the widespread application and inevitable environmental exposure, silver nanoparticles (AgNPs) can be accumulated in various organs. More serious concerns are raised on the biological safety and potential toxicity of AgNPs in the central nervous system (CNS), especially in the hippocampus. This study aimed to investigate the biological effects and the role of PI3K/AKT/mTOR signaling pathway in AgNPs mediated cytotoxicity using the mouse hippocampal neuronal cell line (HT22 cells). AgNPs reduced cell viability and induced membrane leakage in a dose-dependent manner, determined by the MTT and LDH assay. In doses of 25, 50, 100 µg mL-1 for 24 h, AgNPs promoted the excessive production of reactive oxygen species (ROS) and caused the oxidative stress in HT22 cells. AgNPs induced autophagy, determined by the transmission electron microscopy observation, upregulation of LC3 II/I and downregulation of p62 expression levels. The mechanistic investigation showed that the PI3K/AKT/mTOR signaling pathway was activated by phosphorylation, which was enrolled in an AgNP-induced autophagy process. AgNPs could further trigger the apoptosis by upregulation of caspase-3 and Bax and downregulation of Bcl-2 in HT22 cells. These results revealed AgNP-induced cytotoxicity in HT22 cells, which was mediated by autophagy and apoptosis via the PI3K/AKT/mTOR signaling pathway. The study could provide the experimental evidence and explanation for the potential neurotoxicity triggered by AgNPs in vitro.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Nanopartículas Metálicas/toxicidade , Transdução de Sinais/efeitos dos fármacos , Prata/toxicidade , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Serina-Treonina Quinases TOR/metabolismo
11.
J Appl Toxicol ; 40(1): 4-15, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31828819

RESUMO

Micro- and nanoplastics are generated from plastics and have negative impacts on the environment due to their high level of fragmentation. They can originate from various sources such as fragments, fibers and foams. The large proportion of the waste and resistance to degradation means micro- and nanoplastics have become a serious global environmental problem, but there are few studies on their potential toxicity for human health. In this review, we discussed routes of exposure and the potential effects of micro- and nanoplastics to human health. Human beings could mainly be exposed to micro- and nanoplastics orally and by inhalation. The possible toxic effects of plastic particles are due to the potential toxicity of plastics themselves, and their combined toxicity with leachable additives and adsorbed contaminants. The potential risks for human health focused on their gastrointestinal toxicity and liver toxicity. The toxic mechanisms could involve oxidative stress, inflammatory reactions and metabolism disorders. More studies are needed to carry out and explore the potential toxicological mechanisms of micro- and nanoplastics and evaluate the combined toxicity of their adsorbed contaminants.


Assuntos
Ecotoxicologia , Exposição Ambiental/efeitos adversos , Monitoramento Ambiental , Poluentes Ambientais/efeitos adversos , Poluição Ambiental/efeitos adversos , Microplásticos/efeitos adversos , Nanopartículas/efeitos adversos , Animais , Exposição Dietética/efeitos adversos , Cadeia Alimentar , Contaminação de Alimentos , Nível de Saúde , Humanos , Medição de Risco
12.
J Appl Toxicol ; 40(3): 388-402, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31802521

RESUMO

The excellent optical property and relatively low toxicity of CdTe/ZnS core/shell quantum dots (QDs) make them an advanced fluorescent probe in the application of biomedicines, particularly in neuroscience. Thus, it is important to evaluate the biosafety of CdTe/ZnS QDs on the central nervous system (CNS). Our previous studies have suggested that the high possibility of CdTe/ZnS QDs being transported into the brain across the blood-brain barrier resulted in microglial activation and a shift of glycometabolism, but their underlying mechanism remains unclear. In this study, when mice were injected intravenously with CdTe/ZnS QDs through tail veins, the microglial activation, polarized into both M1 phenotype and M2 phenotype, and the neuronal impairment were observed in the hippocampus. Meanwhile, the increased pro- and anti-inflammatory cytokines released from BV2 microglial cells treated with CdTe/ZnS QDs also indicated that QD exposure was capable of inducing microglial activation in vitro. We further demonstrated that the glycolytic shift from oxidative phosphorylation switching into aerobic glycolysis was required in the microglial activation into M1 phenotype induced by CdTe/ZnS QD treatment, which was mediated through the mTOR signaling pathway. The findings, taken together, provide a mechanistic insight regarding the CdTe/ZnS QDs inducing microglial activation and the role of the glycolytic shift in it.


Assuntos
Compostos de Cádmio/toxicidade , Glicólise/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Microglia/efeitos dos fármacos , Pontos Quânticos/toxicidade , Sulfetos/toxicidade , Serina-Treonina Quinases TOR/metabolismo , Telúrio/toxicidade , Compostos de Zinco/toxicidade , Animais , Linhagem Celular , Hipocampo/enzimologia , Hipocampo/ultraestrutura , Masculino , Camundongos Endogâmicos ICR , Microglia/enzimologia , Microglia/ultraestrutura , Estresse Oxidativo/efeitos dos fármacos , Fenótipo , Transdução de Sinais
13.
J Appl Toxicol ; 40(6): 815-831, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31984544

RESUMO

This study evaluated the biodistribution and organ oxidative effects of silver nanoparticles (AgNPs) coated with/without polyvinylpyrrolidone (PVP) (AgNP-20 and AgNP-PVP) in mice; these were administered by gavage at a dose of 10-250 mg/kg body weight per day for 28 days. The results showed that both the AgNPs could induce subacute toxicity and oxidative damage to mice and were mainly accumulated in the liver and spleen and excreted by feces. AgNPs could be absorbed into blood and might cross the blood-brain barrier, and be distributed extensively in mice. The malondialdehyde content in the liver, lungs and kidneys increased in both AgNP groups, while the content of glutathione decreased, and the activity of superoxide dismutase increased at first and then decreased along with the increased doses. Inflammatory pathological changes in the lung and liver at high dose of both AgNPs were consistent with increases in glutamate pyruvic transaminase, glutamate oxaloacetic transaminase and the total protein in serum detection. The Ag content was detected in organs, with the highest content in the liver, followed by spleen, while the Ag content in feces was about 500 times higher than that in urine. AgNP-PVP could induce higher oxidative stress and subacute toxicity than AgNP-20 at the same dose, which might be related to the higher concentrations and more Ag+ ions released in mice after AgNP-PVP exposure. The data from this research provided information on toxicity and biodistribution of AgNPs following gavage administration in mice, and might shed light for future application of AgNPs in daily life.


Assuntos
Nanopartículas Metálicas/toxicidade , Povidona/toxicidade , Compostos de Prata/toxicidade , Administração Oral , Animais , Feminino , Masculino , Nanopartículas Metálicas/administração & dosagem , Camundongos Endogâmicos ICR , Povidona/metabolismo , Compostos de Prata/administração & dosagem , Compostos de Prata/metabolismo , Distribuição Tecidual
14.
J Appl Toxicol ; 39(1): 27-37, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30247756

RESUMO

Gut bacteria are involved in regulating several important physiological functions in the host, and intestinal dysbacteriosis plays an important role in several human diseases, including intestinal, metabolic and autoimmune disorders. Although silver nanoparticles (AgNPs) are increasingly being incorporated into medical and consumer products due to their unique physicochemical properties, studies have indicated their potential to affect adversely the gut bacteria. In this review, we focus on the biotoxicological effects of AgNPs entering the gastrointestinal tract and the relationship of these effects with important nanoscale properties. We discuss in detail the mechanisms underlying the bactericidal toxicity effects of AgNPs and explore the relationships between AgNPs, gut bacteria and disease. Finally, we highlight the need to focus on the negative effects of AgNPs usage to facilitate appropriate development of these particles.


Assuntos
Antibacterianos/toxicidade , Bactérias/efeitos dos fármacos , Microbioma Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/metabolismo , Nanopartículas Metálicas/toxicidade , Prata/metabolismo , Prata/toxicidade
15.
J Appl Toxicol ; 39(6): 908-918, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30701584

RESUMO

With the rapid expansion of human exposure to silver nanoparticles (AgNPs), the genotoxicity screening is critical to the biosafety evaluation of nanosilver. This study assessed DNA damage and chromosomal aberration in the human hepatoma cell line (HepG2) as well as the effects on the micronucleus of bone marrow in mice induced by 20 nm polyvinylpyrrolidone-coated nanosilver (PVP-AgNPs) and 20 nm bare nanosilver (AgNPs). Our results showed that the two types of AgNPs, in doses of 20-160 µg/mL, could cause genetic toxicological changes on HepG2 cells. The DNA damage degree of HepG2 cells in 20 nm AgNPs was higher than that in 20 nm PVP-AgNPs, while the 20 nm PVP-AgNPs caused more serious chromosomal aberration than 20 nm AgNPs. Both kinds of AgNPs caused genetic toxicity in a dose-dependent manner in HepG2 cells. In the micronucleus test on mouse bone marrow cells, in doses of 10, 50 and 250 mg/kg body weight administered orally for 28 days once a day, the two kinds of AgNPs have no obvious inhibitory effect on the mouse bone marrow cells, and the effect of chromosome aberration could be documented at the high dose of 250 mg/kg. These results suggest that AgNPs have genotoxic effects in HepG2 cells and limited effects on bone marrow in mice; both in vitro and in vivo tests could be of great importance on the evaluation of genotoxicity of nanosilver. These findings can provide useful toxicological information that can help to assess genetic toxicity of nanosilver in vitro and in vivo.


Assuntos
Aberrações Cromossômicas/induzido quimicamente , Dano ao DNA , Fígado/efeitos dos fármacos , Nanopartículas Metálicas/toxicidade , Prata/toxicidade , Animais , Apoptose/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Feminino , Células Hep G2 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Testes para Micronúcleos
16.
J Appl Toxicol ; 39(3): 525-539, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30511393

RESUMO

Microglial cells are resident immune cells in the central nervous system. Activation of microglia as induced by CdTe quantum dots (QDs) can trigger damage to neurons. To quantify the intracellular QDs, we monitored the intracellular Cd concentration in the QD-exposed mouse microglial cells (BV-2 cell line). The extent of cell injury at different times correlated with the Cd concentration in cells at that time. In addition to Cd ion detection, we also monitored the intracellular fluorescence of the QDs. More QDs accumulated in the nucleus than in the cytoplasm. Comet assays confirmed that QDs induce DNA damage. However, DNA cannot interact with QDs, so the DNA damage was not caused by CdTe QDs adducts to DNA but by the increase of the Cd ion concentration and the secondary oxidative damage. In addition to DNA damage, biofilm injury and endogenous reduced glutathione depletion were also apparent in QD-exposed BV-2 cells. These changes can be prevented or even reversed by exogenous reduced glutathione administration.


Assuntos
Compostos de Cádmio/toxicidade , Dano ao DNA , Microglia/efeitos dos fármacos , Síndromes Neurotóxicas/etiologia , Pontos Quânticos/toxicidade , Telúrio/toxicidade , Animais , Células Cultivadas , Glutationa/metabolismo , Camundongos , Microglia/metabolismo , Microglia/ultraestrutura , Pontos Quânticos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Espectrofotometria Atômica
17.
Ecotoxicol Environ Saf ; 169: 863-873, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30597786

RESUMO

PM2.5 has implications in cardiovascular adverse events, but the underlying mechanisms are still obscure. The aim of this study is to evaluate miRNA expression in endothelial cells in response to two realistic doses of PM2.5 and to identify the possible gene targets of deregulated miRNAs through microarray profiling and computational technology. As a result, there are 18 differentially expressed miRNAs between 2.5 µg/cm2 group and the control, of which 11 miRNAs are up-regulated and 7 miRNAs are down-regulated. Relative to the control group, 40 miRNAs are significantly changed in 10 µg/cm2 group with 21 miRNAs being upregulated and 19 miRNAs being downregulated. Interestingly, when two PM2.5-treated groups respectively compared with the control, the expressed trends of 12 miRNAs in 2.5 µg/cm2 group are the same as those in 10 µg/cm2 group, with 8 being upregulated and 4 miRNAs being simultaneously downregulated. Gene ontology (GO) analysis shows that the crucial functional categories of miRNA-targeted genes incorporate transcription-related process and intracellular signal transduction. Pathway analysis reveals that endocytosis, FoxO signaling pathway and PI3K-Akt signaling pathway are involved in the PM2.5-caused cardiotoxicity. Further confirmation by RT-qPCR indicates that PM2.5 could induce the down-regulation of hsa-miR-128-3p, hsa-miR-96-5p, hsa-miR-28-5p, hsa-miR-4478 and hsa-miR-6808-5p, which are in accordance with the results of array data. With the comprehensive analysis of mRNAs and miRNAs, a great number of pairs have been identified, suggesting abnormally expressed miRNAs have functions in the cardiotoxicity of PM2.5, and the function may be achieved through the post-transcriptional regulation of certain genes on the related pathways.


Assuntos
Células Endoteliais/efeitos dos fármacos , Poluentes Ambientais/farmacologia , Regulação da Expressão Gênica , MicroRNAs , Tamanho da Partícula , Material Particulado/farmacologia , RNA Mensageiro , Cardiotoxinas , Regulação para Baixo , Exposição Ambiental , Perfilação da Expressão Gênica , Humanos , Fosfatidilinositol 3-Quinases , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais , Regulação para Cima
18.
J Appl Toxicol ; 38(9): 1177-1194, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29722432

RESUMO

Recently, the increasing number of bio-safety assessments on cadmium-containing quantum dots (QDs) suggested that they could lead to detrimental effects on the central nervous system (CNS) of living organisms, but the underlying action mechanisms are still rarely reported. In this study, whole-transcriptome sequencing was performed to analyze the changes in genome-wide gene expression pattern of rat hippocampus after treatments of cadmium telluride (CdTe) QDs with two sizes to understand better the mechanisms of CdTe QDs causing toxic effects in the CNS. We identified 2095 differentially expressed genes (DEGs). Fifty-five DEGs were between the control and 2.2 nm CdTe QDs, 1180 were between the control and 3.5 nm CdTe QDs and 860 were between the two kinds of CdTe QDs. It seemed that the 3.5 nm CdTe QD exposure might elicit severe effects in the rat hippocampus than 2.2 nm CdTe QDs at the transcriptome level. After bioinformatics analysis, we found that most DEG-enriched Gene Ontology subcategories and Kyoto Encyclopedia of Genes and Genomes pathways were related with the immune system process. For example, the Gene Ontology subcategories included immune response, inflammatory response and T-cell proliferation; Kyoto Encyclopedia of Genes and Genomes pathways included NOD/Toll-like receptor signaling pathway, nuclear factor-κB signaling pathway, tumor necrosis factor signaling pathway, natural killer cell-mediated cytotoxicity and T/B-cell receptor signaling pathway. The traditional toxicological examinations confirmed the systemic immune response and CNS inflammation in rats exposed to CdTe QDs. This transcriptome analysis not only revealed the probably molecular mechanisms of CdTe QDs causing neurotoxicity, but also provided references for the further related studies.


Assuntos
Ácido 3-Mercaptopropiônico/toxicidade , Compostos de Cádmio/toxicidade , Perfilação da Expressão Gênica/métodos , Hipocampo/efeitos dos fármacos , Imunidade/efeitos dos fármacos , Pontos Quânticos/toxicidade , Telúrio/toxicidade , Transcriptoma/efeitos dos fármacos , Ácido 3-Mercaptopropiônico/análogos & derivados , Animais , Bases de Dados Genéticas , Redes Reguladoras de Genes/efeitos dos fármacos , Hipocampo/imunologia , Hipocampo/metabolismo , Imunidade/genética , Masculino , Tamanho da Partícula , Ratos Wistar
19.
Ecotoxicol Environ Saf ; 159: 213-220, 2018 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-29753823

RESUMO

Epidemiological studies have illustrated that PM2.5 is closely related to cardiovascular disease (CVD), but underlying toxicological mechanisms are not yet clear. The main purpose of this study is to disclose the potential biological mechanisms responsible for PM2.5-dependent adverse cardiovascular outcomes through the appliance of genome-wide transcription microarray. From results, compared with the control group, there are 97 genes significantly altered in 2.5 µg/cm2 PM2.5 treated group and 440 differentially expressed genes in 10 µg/cm2 group. Of note, when 2.5 µg/cm2 and 10 µg/cm2 group were respectively compared with the control group, 46 significantly altered genes showed a consistent tendency in two treated groups, of which 31 genes were upregulated while 15 genes were meanwhile downregulated. Based on Gene Ontology (GO) annotation, altered genes are mainly gathered in functions of cellular processes and immune regulation. Pathway analysis indicated that TNF signaling pathway, NOD-like receptor (NLRs) signaling pathway, MAPK signaling pathway and gap junction are vital pathways involved in PM2.5-induced toxicity in EA.hy926. Moreover, results from RT-qPCR further corroborated that changed genes are implicated in oxidative stress, inflammation and metabolic disorder. In addition, metabolism of xenobiotics by cytochrome P450 pathway is the critical pathway which may serve as a target to prevent PM2.5-induced CVD. To sum up, our effort provides a fundamental data for further studies regarding mechanisms of PM2.5-induced cardiovascular toxicity on the basis of genome-wide screening.


Assuntos
Poluentes Atmosféricos/toxicidade , Células Endoteliais/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Material Particulado/toxicidade , Linhagem Celular , Células Endoteliais/metabolismo , Perfilação da Expressão Gênica , Humanos , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Proteínas NLR/genética , Análise de Sequência com Séries de Oligonucleotídeos , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/genética
20.
Environ Toxicol ; 32(5): 1530-1538, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-27748997

RESUMO

To investigate the reproductive toxicity and underlying mechanism of nickel nanoparticles (Ni NPs), Caenorhabditis elegans (C. elegans) were treated with/without 1.0, 2.5, and 5.0 µg cm-2 of Ni NPs or nickel microparticles (Ni MPs). Generation time, fertilized egg numbers, spermatide activation and motility were detected. Results indicated, under the same treatment doses, that Ni NPs induced higher reproductive toxicity to C. elegans than Ni MPs. Reproductive toxicities observed in C. elegans included a decrease in brood size, fertilized egg and spermatide activation, but an increase in generation time and out-of-round spermatids. The reproductive toxicity of Ni NPs on C. elegans may be induced by oxidative stress. The reproductive toxicity in C. elegans induced by Ni NPs is consistent with our previous results in the rats. Therefore, C. elegans can be used as an alternative model to detect the early reproductive toxicity of Ni NPs exposure. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 1530-1538, 2017.


Assuntos
Caenorhabditis elegans/efeitos dos fármacos , Caenorhabditis elegans/fisiologia , Fertilização/efeitos dos fármacos , Nanopartículas Metálicas/toxicidade , Níquel/toxicidade , Espermatogênese/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Estudos de Viabilidade , Feminino , Masculino , Modelos Teóricos , Estresse Oxidativo/efeitos dos fármacos , Reprodução/efeitos dos fármacos , Motilidade dos Espermatozoides/efeitos dos fármacos , Testes de Toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA