Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Chemosphere ; 331: 138817, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37127200

ABSTRACT

Tris(2-chloroisopropyl) phosphate (TCPP) and Tris(2-chloroethyl) phosphate (TCEP) are the widely used organophosphorus flame retardants indoors and easily accessible to the eyes as the common adhesive components of dust and particle matter, however, hardly any evidence has demonstrated their corneal toxicity. In this study, the adverse effects of TCPP, TCEP, and TCPP + TCEP exposure on human corneal epithelial cells (HCECs) were investigated. The cell viability and morphology, intracellular reactive oxygen species (ROS), cell cycle, and the expressions of cell cycle and pyroptosis-related genes were assessed to explain the underlying mechanisms. Compared to individual exposure, co-exposure to TCPP20+TCEP20 showed higher cytotoxicity with a sharp decrease of >30% in viability and more serious oxidative damage by increasing ROS production to 110.92% compared to the control group. Furthermore, the cell cycle arrested at the S phase (36.20%) was observed after combined treatment, evidenced by the upregulation of cyclin D1, CDK2, CDK4, CDK6, p21, and p27. Interestingly, pyroptosis-related genes GSDMD, Caspase-1, NLRP3, IL-1ß, IL-18, NLRP1, and NLRC4 expressions were promoted with cell swelling and glowing morphology. Oxidative stress and cell cycle arrest probably acted as a key role in TCPP20+TCEP20-induced cytotoxicity and pyroptosis in HCECs. Our results suggested that TCPP20+TCEP20 co-exposure induced severer corneal damage, further illustrating its significance in estimating indoor health hazards to humans.


Subject(s)
Flame Retardants , Pyroptosis , Humans , Reactive Oxygen Species/metabolism , Epithelial Cells/metabolism , Oxidative Stress , Cell Cycle Checkpoints , Phosphates/metabolism , Flame Retardants/toxicity
2.
Int J Mol Sci ; 23(22)2022 Nov 18.
Article in English | MEDLINE | ID: mdl-36430782

ABSTRACT

Tris (1-chloro-2-propyl) phosphate (TCPP) is one of the most frequently detected organophosphorus flames in the environment. Continuous daily exposure to TCPP may harm human skin. However, little is known about the adverse effects of TCPP on human skin. In this study, we first evaluated the detrimental effects and tried to uncover the underlying mechanisms of TCPP on human skin keratinocytes (HaCaT) after 24 h exposure. We found that TCPP caused a concentration-dependent decrease in HaCaT cell viability after exposure to 1.56-400 µg/mL for 24 h, with an IC50 of 275 µg/mL. TCPP also promoted the generation of intracellular reactive oxygen species (ROS) and triggered DNA damage, evidenced by an increase of phosphorylated histone H2A.X (γH2A.X) in the nucleus. Furthermore, the cell cycle was arrested at the G1 phase at 100 µg/mL by upregulation of the mRNA expression of p53 and p21 and downregulation of cyclin D1 and CDK4 expression. Additionally, both the senescence-associated-ß-galactosidase activity and related proinflammatory cytokine IL-1ß and IL-6 were elevated, indicating that TCPP exposure caused cellular senescence may be through the p53-dependent DNA damage signal pathway in HaCaT cells. Taken together, our data suggest that flame-retardant exposure may be a key precipitating factor for human skin aging.


Subject(s)
Flame Retardants , Skin Aging , Humans , Cellular Senescence , Flame Retardants/toxicity , Keratinocytes/metabolism , Organophosphorus Compounds/toxicity , Organophosphorus Compounds/metabolism , Tumor Suppressor Protein p53/metabolism
3.
Int J Mol Sci ; 23(19)2022 Oct 04.
Article in English | MEDLINE | ID: mdl-36233064

ABSTRACT

Cadmium (Cd) is one of the toxic heavy metals found widely in the environment. Skin is an important target organ of Cd exposure. However, the adverse effects of Cd on human skin are still not well known. In this study, normal human skin keratinocytes (HaCaT cells) were studied for changes in cell viability, morphology, DNA damage, cycle, apoptosis, and the expression of endoplasmic reticulum (ER) stress-related genes (XBP-1, BiP, ATF-4, and CHOP) after exposure to Cd for 24 h. We found that Cd decreased cell viability in a concentration-dependent manner, with a median lethal concentration (LC50) of 11 µM. DNA damage induction was evidenced by upregulation of the level of γ-H2AX. Furthermore, Cd induced G0/G1 phase cell cycle arrest and apoptosis in a dose-dependent manner and upregulated the mRNA levels of ER stress biomarker genes (XBP-1, BiP, ATF4, and CHOP). Taken together, our results showed that Cd induced cytotoxicity and DNA damage in HaCaT cells, eventually resulting in cell cycle arrest in the G0/G1 phase and apoptosis. In addition, ER stress may be involved in Cd-induced HaCaT apoptosis. Our data imply the importance of reducing Cd pollution in the environment to reduce its adverse impacts on human skin.


Subject(s)
Cadmium , Endoplasmic Reticulum Stress , Apoptosis , Cadmium/toxicity , Humans , Keratinocytes , RNA, Messenger
4.
Sci Total Environ ; 835: 155544, 2022 Aug 20.
Article in English | MEDLINE | ID: mdl-35489519

ABSTRACT

The dermal exposure of heavy metals in contaminated urban soils poses huge environmental health risks globally. However, their dermal bioaccessibility and adverse effects on human skin cells were not fully understood. In this study, we measured the total and dermal bioaccessibility of Cr, As, Cd, Pb, and Cu in four selected urban soil samples from Kunming, Yunnan, China, and evaluated the cellular responses of these bioaccessible extracts on human keratinocytes (HaCaT). Among all the metals, only As in Soil-3 (S3) exceeded Chinese risk screening and Yunnan background values at 38.2 mg/kg. The average concentrations of Cr, As, Cd, Pb, and Cu in all soil samples were 47.79, 15.50, 3.11, 104.27, and 180.29 mg/kg respectively. Although relatively high concentrations of heavy metals were detected in soil samples, the highest dermal bioaccessibility of Cd was 3.57% with others' being lower than 1%. The bioaccessible dermal-absorbed doses (DADs) of Cr, As, Cd, Pb, and Cu from soils reflected acceptable health risks since all DADs were below the corresponding derived dermal reference values. However, the toxic data showed the extracts of S3 and S4 presented certain cytotoxicity in HaCaT cells, indicating the existing models based on dermal bioaccessibility and DADs may be not accurate enough to assess their human health risk. Taken together, the human health risk assessment should be modified by taking their skin cytotoxicity into account.


Subject(s)
Metals, Heavy , Soil Pollutants , Cadmium , China , Environmental Monitoring , Humans , Lead , Metals, Heavy/analysis , Metals, Heavy/toxicity , Risk Assessment , Soil , Soil Pollutants/analysis , Soil Pollutants/toxicity
5.
Chemosphere ; 292: 133361, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34929273

ABSTRACT

E-wastes release toxic metals including Cd, Cu, Ni, Pb and Zn into nearby soils during dismantling process. However, their adverse effects and the associated mechanisms on human intestinal epithelium are poorly understood. In this study, their toxic effects on human colonic epithelial cells Caco-2 and the underlying mechanisms were assessed basing on three soils from Wenling e-waste dismantling site. Since soil-extractable metals are more available for gastrointestinal absorption, we used phosphate buffer saline solution to extract metals at solid to liquid ratio of 1:2. Among metals, total Cd and Ni exceeded the risk screening values in three soils, being 3.8-8.8 and 42.4-155 mg/kg. Furthermore, high extractable-metals at 5.9, 1.9, and 0.87 mg/kg Cd (20-67%) and 4.6, 6.4, and 12.4 mg/kg Ni (3.6-29%) were observed for Soil-1, -2 and -3, respectively. All three extracts triggered cytotoxicity, with Soil-2 showing the strongest inhibition of cell viability. Higher production of reactive oxygen species and stronger inhibition of antioxidant enzymes SOD1 and CAT were observed in Soil-2 and -3. Upregulation of proinflammatory mediators (IL-1ß, IL-8 and TNF-α) and apoptosis-regulatory genes (GADD45α, Caspase-3, and Caspase-8) were observed. Our data suggest that soil extracts induced cytotoxicity, oxidative damage, inflammatory response, and cell apoptosis in Caco-2 cells, indicating soil ingestion from e-waste dismantling site may adversely impact human health.


Subject(s)
Electronic Waste , Metals, Heavy , Soil Pollutants , Caco-2 Cells , Cadmium/toxicity , China , Environmental Monitoring , Humans , Metals, Heavy/analysis , Metals, Heavy/toxicity , Risk Assessment , Soil , Soil Pollutants/analysis , Soil Pollutants/toxicity
6.
Sci Total Environ ; 756: 143951, 2021 Feb 20.
Article in English | MEDLINE | ID: mdl-33261865

ABSTRACT

Cadmium (Cd) and copper (Cu) are widely present in foods. However, their adverse effects on human gastric epithelium are not fully understood. Here, human gastric epithelial cells (SGC-7901) were employed to study the toxicity and associated mechanisms of Cd + Cu co-exposure. Their effects on cell viability, morphology, oxidative damage, cell cycle, apoptosis, and the mRNA levels of antioxidases and cell cycle regulatory genes were investigated. Co-exposure to Cd (5 µM)/Cu (10 µM) induced >40% cell viability loss, whereas little effect on cell viability at <10 µM Cd or 40 µM Cu. Compared to individual exposure, co-exposure induced greater oxidative damage by elevating ROS (3.5 folds), malondialdehyde (2.3 folds) and expression of SOD1 and HO-1 besides inhibiting CAT, GPX1 and Nrf2. A marked S cell-cycle arrest was observed in co-exposure, evidenced by more cells staying in the S phase (36%), up-regulation of cyclins-dependent kinase (CDK4) and CDKs inhibitor (p21) and down-regulation of CDK2, CDK6 and p27. Furthermore, higher apoptosis (22%) with floated and round cells occurred in co-exposure group. Our data implicate the cytotoxicity of Cd + Cu co-exposure was higher than individual exposure, and individual assessment would underestimate their potential health risk. Oxidative stress and cell cycle arrest possibly played a role in Cd + Cu induced toxicity and apoptosis in SGC-7901 cells. Our data suggest the importance to reduce Cd in foods to decrease its adverse impacts on human digestive system.


Subject(s)
Cadmium , Oxidative Stress , Apoptosis , Cadmium/toxicity , Cell Cycle Checkpoints , Epithelial Cells , Humans
7.
Mol Med Rep ; 15(4): 1981-1988, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28260032

ABSTRACT

Slit homolog 2 (Slit2) is distributed in various tissues and participates in numerous cellular processes; however, the role of Slit2 in the regulation of angiogenesis remains controversial, since it has previously been reported to exert proangiogenic and antiangiogenic activities. The present study aimed to investigate the effects of Slit2 on vascular endothelial cell proliferation and migration in vitro, and to reveal the possible underlying signaling pathway. Aortic endothelial cells were isolated from Sprague Dawley rats and cultured. Cell proliferation assay, cell migration assay, immunocytochemistry and small interfering RNA transfection were subsequently performed. The results demonstrated that exogenous Slit2 administration markedly suppressed TNF­α­induced endothelial cell proliferation and migration in vitro. In addition, TNF­α application upregulated the protein expression levels of vascular endothelial growth factor (VEGF) and Notch in RAECs, whereas Slit2 administration downregulated VEGF and Notch expression in RAECs cultured in TNF­α conditioned medium. Further studies indicated that knockdown of VEGF suppressed the effects of TNF­α on the induction of RAEC proliferation and migration. VEGF knockdown­induced inhibition of RAEC proliferation and migration in TNF­α conditioned medium was also achieved without Slit2 administration. Furthermore, VEGF knockdown markedly decreased Notch1 and Notch2 expression. These results indicated that Slit2 suppresses TNF­α­induced vascular endothelial cell proliferation and migration in vitro by inhibiting the VEGF­Notch signaling pathway. Therefore, Slit2 may inhibit the proliferation and migration of endothelial cells during vascular development.


Subject(s)
Cell Movement , Cell Proliferation , Endothelial Cells/cytology , Intercellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Notch/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Animals , Aorta/cytology , Cells, Cultured , Endothelial Cells/metabolism , Rats, Sprague-Dawley , Receptors, Notch/genetics , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...