Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Neurochem Res ; 49(9): 2453-2468, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38850437

RESUMEN

Tri(1,3-dichloro-2-propyl)phosphate (TDCPP) is one of the most widely used organophosphorus flame retardants in consumer products. TDCPP has been confirmed to be neurotoxic, but its mechanism has not been clarified and may be related to mitophagy. AMBRA1 can promote neurological autophagy, but whether AMBRA1 is involved in the mechanism of TDCPP-induced neurotoxicity has not been elucidated. In this study, the optimal neuronal damage model was established by exposing mice hippocampal neurons to TDCPP. Furthermore, on the basis of this model, siRNA was used to knock down AMBRA1. Combined with qRT-PCR and Western blot techniques, we identified AMBRA1-mediated mitophagy-induced neuronal damage in vitro mechanism. The experimental results indicated that TDCPP treatment for 24 h led to a decrease in the cell viability of mouse hippocampal neurons, causing neuronal damage. Meanwhile, TDCPP exposure increased autophagy marker proteins p62 and LC3B, and down-regulated mitochondrial DNA ND1 damage and TOMM20 protein, suggesting that TDCPP exposure promoted mitophagy. In addition, TDCPP exposure led to changes in the expression of AMBRA1 and the key factors of mitophagy, FUNDC1, PINK1, and PARKIN, whereas mitophagy was inhibited after knockdown of AMBRA1. The research results indicated that exposure to TDCPP induced neuronal damage and promoted mitophagy. The mechanism may be that AMBRA1 promoted mitophagy in neuronal cells through the PARKIN-dependent/non-dependent pathway. This study revealed the toxic effects of TDCPP on the nervous system and its potential molecular mechanisms, which provided important clues for further understanding the mechanism of action of AMBAR1-mediated mitophagy.


Asunto(s)
Hipocampo , Mitofagia , Neuronas , Animales , Mitofagia/efectos de los fármacos , Mitofagia/fisiología , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Ratones , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Compuestos Organofosforados/toxicidad , Retardadores de Llama/toxicidad , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales
2.
Environ Pollut ; 347: 123740, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38462198

RESUMEN

Tris (1,3-dichloro-2-propyl) phosphate (TDCPP), a halogen-containing phosphorus flame retardant, is widely used and has been shown to possess health risks to humans. The sustained release of artificial nanomaterials into the environment increases the toxicological risks of their coexisting pollutants. Nanomaterials may seriously change the environmental behavior and fate of pollutants. In this study, we investigated this combined toxicity and the potential mechanisms of toxicity of TDCPP and titanium dioxide nanoparticles (TiO2 NPs) aggregates on human neuroblastoma SH-SY5Y cells. TDCPP and TiO2 NPs aggregates were exposed in various concentration combinations, revealing that TDCPP (25 µg/mL) reduced cell viability, while synergistic exposure to TiO2 NPs aggregates exacerbated cytotoxicity. This combined exposure also disrupted mitochondrial function, leading to dysregulation in the expression of mitochondrial fission proteins (DRP1 and FIS1) and fusion proteins (OPA1 and MFN1). Consequently, excessive mitochondrial fission occurred, facilitating the translocation of cytochrome C from mitochondria to activate apoptotic signaling pathways. Furthermore, exposure of the combination of TDCPP and TiO2 NPs aggregates activated upstream mitochondrial autophagy but disrupted downstream Parkin recruitment to damaged mitochondria, preventing autophagosome-lysosome fusion and thereby disrupting mitochondrial autophagy. Altogether, our findings suggest that TDCPP and TiO2 NPs aggregates may stimulate apoptosis in neuronal SH-SY5Y cells by inducing mitochondrial hyperfission and inhibiting mitochondrial autophagy.


Asunto(s)
Contaminantes Ambientales , Neuroblastoma , Humanos , Mitofagia , Neuroblastoma/metabolismo , Dinámicas Mitocondriales , Apoptosis
3.
Heliyon ; 10(3): e25554, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38327441

RESUMEN

Tris (1,3-dichloro-2-propyl) phosphate (TDCPP) is a growing concern and may be a potential risk to marine environmental health due to its widespread usage and distribution. However, the toxic effects of TDCPP on cardiac development in marine fish have not been reported. In this study, Oryzias melastigma embryos were exposed to TDCPP at doses of 0, 0.04, 0.4, 4 and 40 µg/L from early embryogenesis to 10 days postfertilization (dpf). Then, the heart rate and sinus venosus-bulbus arteriosus (SV-BA) distance of the exposed embryos were measured at 5, 6, 8 and 10 dpf. Furthermore, alterations in the mRNA levels of the genes encoding cyclooxygenase-2 (COX-2), bone morphogenetic protein 4 (BMP4), fibroblast growth factor 8 (FGF8), and GATA-binding protein 4 (GATA4) were evaluated at 5, 6, 8 and 10 dpf. We found that the heart rate significantly increased in all TDCPP exposure groups at 10 dpf. The SV-BA distance significantly decreased in all TDCPP exposure groups at all developmental stages (except for the 0.4 µg/L group at 5 dpf and the 4 µg/L group at 10 dpf). The mRNA expression of COX-2 was downregulated at 5 dpf, BMP4 was downregulated at 5 and 6 dpf, FGF8 was downregulated at 5, 6 and 8 dpf, GATA4 was downregulated at 8 dpf, and GATA4 was upregulated at 10 dpf. These results indicate that the changes in heart rate and SV-BA distance might be accompanied by disturbances in the four genes involved in cardiac development. Our findings will help to illustrate the possible cardiac toxic effects of marine fish exposed to TDCPP.

4.
Ecotoxicol Environ Saf ; 265: 115535, 2023 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-37776817

RESUMEN

The objective of this study was to assess the thyroid hormone disruption and reproductive dysfunction effects of the bioaccumulation and rate of mechanism in zebrafish exposed to tris(1,3-dichloro-2-propyl) phosphate (TDCPP), with stress responsiveness. The fish were exposed to test concentrations of TDCPP (0, 0.06, 0.3, 1.5 µg/mL) for 21 days, in accordance with no observed adverse effect level (i.e., < EC10) for zebrafish embryos. The bioaccumulation of TDCPP was found to be significantly higher in female zebrafish, while the metabolic rate was significantly higher in male zebrafish at all concentrations studied. The thyroid hormone (triiodothyronine [T3] and thyroxine [T4]) levels and sex steroid (i.e., estrogen, androgen, and progesterone) levels were significantly increased only in female zebrafish exposed to TDCPP, and no significant difference was observed in male zebrafish, although their cortisol levels increased. The response to TDCPP can, therefore, be considered sex-specific. The results of this study demonstrate for the first time, that the different response in the bioaccumulation and metabolic rate of TDCPP in males and females. The results also indicate that TDCPP alters thyroid hormone levels, furthermore, as steroidogenesis is related to reproductive function with differing response in males and females. TDCPP can be assumed to exert reproductive toxicity via disruption of thyroid and steroid synthesis through a slow metabolic rate in the whole body after exposure. Consequently, our proposed methodological approach to assess the interactions of thyroid and steroid biosynthesis and metabolic rate of TDCPP with reproductive toxicity will serve a testing strategy to examine the adverse outcomes of emerging environmental chemicals.

5.
Sci Total Environ ; 894: 164823, 2023 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-37343856

RESUMEN

Organophosphorus flame retardants (OPFRs) are now drawing the public's attention due to their potential toxicity. Given that contaminated food may result in the ingestion of OPFRs to the human intestine, further investigation is required to determine the potential adverse effects of these compounds on human intestinal health. The present study aimed to comprehensively assess the effect of tris(1,3-dichloro-2-propyl) phosphate (TDCPP), a typical OPFR, on human intestinal health by evaluating both intestinal flora and human cell Caco-2. Based on the results, TDCPP exposure altered the composition of intestinal flora and increased the proportion of pathogenic bacteria. PICRUSt2 analysis revealed that certain pathways were affected by TDCPP, and the resulting metabolic disorders might cause health problems. Orthologous genes of glutathione S-transferase and multidrug efflux system were up-regulated, demonstrating that the bacteria resisted TDCPP to maintain their vitality. Compared to the other two OPFRs, TDCPP induced greater cytotoxicity, and the results were consistent with the dose-effect relationship. Three OPFRs, especially TDCPP, caused the release of lactate dehydrogenase, accumulation of ROS, decline in mitochondrial membrane potential and increase in intracellular Ca2+, which could consequently induce cell death. The simultaneous effects of TDCPP on both intestinal cells and intestinal flora are likely to engender more severe intestinal health issues.


Asunto(s)
Retardadores de Llama , Microbioma Gastrointestinal , Humanos , Fosfatos/toxicidad , Organofosfatos/toxicidad , Organofosfatos/metabolismo , Compuestos Organofosforados/toxicidad , Células CACO-2 , Retardadores de Llama/toxicidad , Retardadores de Llama/metabolismo , Intestinos
6.
Aquat Toxicol ; 258: 106510, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37003012

RESUMEN

Tri(1,3-dichloropropyl) phosphate (TDCPP) is widespread in the environment as a typical thyroid hormone-disrupting chemical. Here, we aimed to explore the toxicological mechanisms of the thyroid hormone-disrupting effects induced by TDCPP in zebrafish embryos/larvae using multi-omics analysis. The results showed that TDCPP (400 and 600 µg/L) induced phenotypic alteration and thyroid hormone imbalance in zebrafish larvae. It resulted in behavioral abnormalities during zebrafish embryonic development, suggesting that this chemical might exhibit neurodevelopmental toxicity. Transcriptomic and proteomic analysis provided consistent evidence at the gene and protein levels that neurodevelopmental disorders were significantly enhanced by TDCPP exposure (p < 0.05). Additionally, multi-omics data indicated that membrane thyroid hormone receptor (mTR)-mediated non-genomic pathways, including cell communication (ECM-receptor interactions, focal adhesion, etc.) and signal transduction pathways (MAPK signaling pathway, calcium signaling pathway, neuroactive ligand-receptor interaction pathway, etc.), were significantly disturbed (p < 0.05) and might contribute to the neurodevelopmental toxicity induced by TDCPP. Therefore, behavioral abnormalities and neurodevelopmental disorders might be important phenotypic characteristics of TDCPP-induced thyroid hormone disruption, and mTR-mediated non-genomic networks might participate in the disruptive effects of this chemical. This study provides new insights into the toxicological mechanisms of TDCPP-induced thyroid hormone disruption and proposes a theoretical basis for risk management of this chemical.


Asunto(s)
Contaminantes Químicos del Agua , Pez Cebra , Animales , Pez Cebra/metabolismo , Compuestos Organofosforados/toxicidad , Multiómica , Proteómica , Contaminantes Químicos del Agua/toxicidad , Hormonas Tiroideas/metabolismo , Fosfatos/metabolismo
7.
Toxics ; 11(3)2023 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-36976996

RESUMEN

Tris(1,3-dichloro-2-propyl) phosphate (TDCPP) is an organophosphorus flame retardant that has been utilized in recent years as a primary replacement for polybrominated diphenyl ethers (PBDEs) in a wide variety of fire-sensitive applications. However, the impact of TDCPP on the immune system has not been fully determined. As the largest secondary immune organ in the body, the spleen is considered to be an important study endpoint for determining immune defects in the body. The aim of this study is to investigate the effect of TDCPP toxicity on the spleen and its possible molecular mechanisms. In this study, for 28 consecutive days, TDCPP was administered intragastrically (i.g), and we assessed the general condition of mice by evaluating their 24 h water and food intake. Pathological changes in spleen tissues were also evaluated at the end of the 28-day exposure. To measure the TDCPP-induced inflammatory response in the spleen and its consequences, the expression of the critical players in the NF-κB pathway and mitochondrial apoptosis were detected. Lastly, RNA-seq was performed to identify the crucial signaling pathways of TDCPP-induced splenic injury. The results showed that TDCPP intragastric exposure triggered an inflammatory response in the spleen, likely through activating the NF-κB/IFN-γ/TNF-α/IL-1ß pathway. TDCPP also led to mitochondrial-related apoptosis in the spleen. Further RNA-seq analysis suggested that the TDCPP-mediated immunosuppressive effect is associated with the inhibition of chemokines and the expression of their receptor genes in the cytokine-cytokine receptor interaction pathway, including four genes of the CC subfamily, four genes of the CXC subfamily, and one gene of the C subfamily. Taken together, the present study identifies the sub-chronic splenic toxicity of TDCPP and provides insights on the potential mechanisms of TDCPP-induced splenic injury and immune suppression.

8.
Toxicol In Vitro ; 87: 105523, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36427757

RESUMEN

Since 2004, some legacy flame retardants (FRs) were restricted or removed from the European markets due to their concern on human health. Both organophosphorus FRs (OPFRs) and novel brominated FRs (NBFRs) have replaced them because they are presumably safer and less persistent emerging FRs (EFRs) and their exposure is currently occurring in indoor environments at high levels. Little is known about the neurotoxic potential risk of these EFRs in humans. The present study was aimed at assessing the acute neurotoxicity potential of Tris(1, 3-dichloro-2-propyl)phosphate (TDCPP), triphenyl phosphate (TPhP), Bis(2-ethylhexyl)tetrabromophthalate (BEH-TEBP) and 2-ethylhexyl-2,3,4,5-tetrabromobenzoate (EH-TBB) on human neuroblastoma cells (SH-SY5Y). SH-SY5Y were exposed to these EFRs at low concentrations -ranging 2.5-20 µM. during 2-24 h. We investigated viability, mitochondrial function, oxidative stress, inflammatory response, as well as neural plasticity and development. The results have demonstrated that selected EFRs (TDCPP, TPhP, EH-TBB and BEH-TBP) did not impair neural function on SH-SY5Y as acute response. To the best of our knowledge, this has been the first study focused on evaluating the neural affection of TPhP on SH-SY5Y cells and of EH-TBB and BEH-TBP on neural cells. We also assessed for the first time almost all endpoints after FR exposure on neural cell lines.


Asunto(s)
Retardadores de Llama , Neuroblastoma , Humanos , Monitoreo del Ambiente , Retardadores de Llama/toxicidad , Polvo/análisis , Organofosfatos/toxicidad , Compuestos Organofosforados/toxicidad , Éteres Difenilos Halogenados
9.
Toxicol Lett ; 374: 31-39, 2023 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-36493961

RESUMEN

Tris(1,3-dichloro-2-propyl) phosphate (TDCPP) is an organophosphate flame retardant. The primary TDCPP metabolite, bis(1,3-dichloro-2-propyl) phosphate (BDCPP), is detectable in the urine of over 90 % of Americans. Epidemiological studies show sex-specific associations between urinary BDCPP levels and metabolic syndrome, which is an established risk factor for type 2 diabetes, heart disease, and stroke. We used a mouse model to determine whether TDCPP exposure disrupts glucose homeostasis. Six-week old male and female C57BL/6J mice were given ad libitum access to diets containing vehicle (0.1 % DMSO) and TDCPP resulting in the following treatment groups: 0 mg/kg/day, 0.02 mg/kg/day, 1 mg/kg/day, or 100 mg/kg/day. After being on the experimental diet for five weeks without interruption, body composition was analyzed, glucose and insulin tolerance tests were performed, and fasting glucose and insulin levels were quantified. TDCPP at 100 mg/kg/day caused male sex-specific adiposity, fasting hyperglycemia, and insulin resistance. TDCPP-induced modulation of nuclear receptor activation was investigated using an in vitro screen to identify potential mechanisms of metabolic disruption. TDCPP activated farnesoid X receptor (FXR) and pregnane X receptor (PXR), and inhibited the androgen receptor (AR). PXR target genes, but not FXR target genes, were upregulated in livers from mice exposed to 100 mg TDCPP/kg/day. Interestingly, PXR target genes were differentially expressed in livers from both males and females. It remains to be determined whether TDCPP-induced metabolic disruption occurs via modulation of nuclear receptor activity. Taken together, these studies build upon the association of TDCPP exposure and metabolic syndrome in humans by identifying sex-specific effects of TDCPP on glucose homeostasis in mice.


Asunto(s)
Retardadores de Llama , Síndrome Metabólico , Compuestos Organofosforados , Animales , Femenino , Humanos , Masculino , Ratones , Diabetes Mellitus Tipo 2/epidemiología , Retardadores de Llama/metabolismo , Retardadores de Llama/toxicidad , Síndrome Metabólico/inducido químicamente , Síndrome Metabólico/epidemiología , Síndrome Metabólico/orina , Ratones Endogámicos C57BL , Compuestos Organofosforados/metabolismo , Compuestos Organofosforados/toxicidad , Compuestos Organofosforados/orina , Receptores Citoplasmáticos y Nucleares/metabolismo , Resistencia a la Insulina
10.
Chemosphere ; 308(Pt 2): 136345, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36087716

RESUMEN

Tris (1,3-dichloro-2-propyl) phosphate (TDCPP) has neurotoxicity, but its mechanism remains unclear. Evidence recently showed that ferroptosis might be associated with TDCPP-induced neurotoxicity. To explore the role and underlying mechanism of ferroptosis in TDCPP-induced neurotoxicity, the occurrence of ferroptosis was examined in mice and PC12 cells upon TDCPP exposure. The mechanism of TDCPP-induced ferroptosis was clarified in vitro combined with the RNA sequencing assay. The in vivo results showed that orally TDCPP exposure (100 mg/kg, 30 d) inhibited the learning and memory ability of mice, reduced hippocampus neurons, induced malondialdehyde (MDA) accumulation, and decreased glutathione (GSH) and superoxide dismutase (SOD) levels in the hippocampus. Moreover, TDCPP exposure (100 mg/kg, 30 d) altered the ferroptosis and autophagy-related protein abundances in the hippocampus. The in vitro results showed that TDCPP exposure (0, 5, 20, 50, 100, and 200 µM) for 24 h induced dose-dependent cell death in PC12 cells, and the cell death was ameliorated by the co-treatment with ferrostatin-1 (1 µM, 24 h). Similarly, TDCPP exposure (0, 50, 100, and 200 µM) for 24 h increased the levels of MDA and LPO, but decreased the reduced GSH in PC12 cells. Furthermore, TDCPP exposure (0, 50, 100, and 200 µM) for 24 h altered the ferroptosis and autophagy-related protein abundances in PC12 cells. The RNA-sequencing revealed that TDCPP exposure (100 µM, 24 h) induced mitophagy activation in SH-SY5Y cells. Meanwhile, the in vitro experiments confirmed that TDCPP exposure (0, 50, 100, and 200 µM) for 24 h increased abundances of mitophagy-related protein phosphatase and tensin homolog induced kinase 1(PINK1), Parkinson protein 2 E3 ubiquitin-protein ligase (PARKIN), inositol 1,4,5-trisphosphate receptor type 1 (IP3R1), and voltage-dependent anion channel 1 (VDAC1) in PC12 cells. Moreover, TDCPP treatment (100 µM, 24 h) increased the mitochondrial recruitment of PARKIN, decreased the mitochondrial membrane potential (MMP) level, and increased the Fe2+ level in mitochondria. In addition, decreased ATP levels and increased reactive oxygen species (ROS) levels were observed in PC12 cells upon TDCPP exposure (0, 50, 100, and 200 µM) for 24 h. In summary, ferroptosis was associated with TDCPP-induced neurotoxicity, and the mechanism might be related to PINK1/PARKIN-mediated mitophagy initiated by mitochondrial damage.


Asunto(s)
Ferroptosis , Retardadores de Llama , Neuroblastoma , Síndromes de Neurotoxicidad , Adenosina Trifosfato , Animales , Proteínas Relacionadas con la Autofagia , Glutatión/metabolismo , Humanos , Receptores de Inositol 1,4,5-Trifosfato , Malondialdehído , Ratones , Mitofagia/fisiología , Compuestos Organofosforados , Fosfatos/metabolismo , Proteínas Quinasas/metabolismo , ARN , Ratas , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo , Tensinas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Canal Aniónico 1 Dependiente del Voltaje
11.
Food Chem Toxicol ; 169: 113432, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36115506

RESUMEN

Environmental monitoring data have indicated that three chlorinated organophosphorus flame retardants (Cl-OPFRs), including tris(2-chloroethyl)-phosphate (TCEP), tris(2-chloropropyl)-phosphate (TCPP), and tris(1,3-dichloro-2-propyl)-phosphate (TDCPP) are the predominant chemicals in various environmental matrices and exhibit reproductive endocrine disrupting activities. Currently, mitochondrial abnormality is a new paradigm for evaluating chemical-mediated cell dysfunction. However, a comprehensive correlation between these two aspects of Cl-OPFRs remains unclear. In this research, the effects of TCEP, TCPP, and TDCPP on progesterone production and mitochondrial impairment were investigated by using mouse Leydig tumor cells (mLTC-1). The half maximal inhibitory concentration (IC50) values at 48 h exposure indicated that the rank order of anti-androgenic activity was TDCPP > TCPP. Whereas, TCEP exhibited elevation of progesterone production. At concentrations close to IC50 of progesterone production by TCPP and TDCPP, the elevation of intracellular reactive oxygen species (ROS), depletion of mitochondrial membrane potential (MMP), reduction of cellular adenosine triphosphate (ATP) content, and alteration of mitochondrial structures was observed. In addition, the expression of main genes related to progesterone synthesis was dramatically down-regulated by TCPP and TDCPP treatments. These results imply that the inhibition effect of TCPP and TDCPP on progesterone production might be related to mitochondrial damage and down-regulated steroidogenic genes.


Asunto(s)
Monitoreo del Ambiente , Retardadores de Llama , Mitocondrias , Organofosfatos , Fosfinas , Progesterona , Animales , Ratones , Adenosina Trifosfato/metabolismo , Retardadores de Llama/toxicidad , Organofosfatos/toxicidad , Fosfinas/toxicidad , Progesterona/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Tumor de Células de Leydig , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Monitoreo del Ambiente/métodos
12.
Chemosphere ; 305: 135485, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35764118

RESUMEN

Tris (1,3-dichloro-2-propyl) phosphate (TDCPP), a chlorinated organophosphate flame retardants(OPFRs), is widely used in a range of plastic foams, resins, and latexes. It can be detected in human tissues, including urine, and milk. Recent research has suggested that TDCPP has neurotoxic, reproductive, and potentially carcinogenic. In our study, we proposed a novel method for predicting the gene associated with tumor-compound interactions. We firstly used The Comparative Toxicogenomics Database (CTD) and downloaded potentially interactive genes about TDCPP in renal carcinoma. Gene expression data and the corresponding clinical information of the Kidney renal clear cell cancer (KIRC) patients were obtained from The Cancer Genome Atlas database (TCGA). Data from normal people in The Genotype-Tissue Expression (GTEx) databases was used to supplement the calculations. After being predicted by PharmMapper database, and validated by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes, 25 genes were selected to construct protein-protein interaction network analysis. The prognostic value of these genes was evaluated with Kaplan-Meier analysis, and four interactive genes were selected. Gene set variation analysis and drug-target binding prediction proved the hub gene has a potential relationship with renal clear cell carcinoma. We then used the ChEA3 (Chip-X Enrichment Analysis, Version 3) database to predict the upstream of these interactive genes. Molecular docking was used to predict the binding of these transcription factors to TDCPP and interactive genes to TDCPP. Moreover, in cell lines and in vivo experiments demonstrated the cancer-promoting effect of TDCPP. The expression of the interactive genes was verified by qPCR and Western blot. Combining binding energy and qPCR results, we choose EPAS1 to verify its function in renal carcinoma cell lines. Our study provides a novel method to predict the potential interactive genes between TDCPP and renal cancer, which may reveal potential targets for the treatment and prevention of diseases.


Asunto(s)
Carcinoma de Células Renales , Retardadores de Llama , Neoplasias Renales , Carcinoma de Células Renales/inducido químicamente , Carcinoma de Células Renales/genética , Retardadores de Llama/análisis , Humanos , Neoplasias Renales/inducido químicamente , Neoplasias Renales/genética , Simulación del Acoplamiento Molecular , Organofosfatos/metabolismo , Organofosfatos/toxicidad , Compuestos Organofosforados , Factores de Riesgo
13.
J Appl Toxicol ; 42(8): 1385-1395, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35172387

RESUMEN

We compared the influence of thyroid hormone-disrupting chemicals (heptafluorobutanoic acid, PFBA and tris[1,3-dichloro-2-propyl] phosphate, TDCPP) and thyroid hormone (3,3',5-triiodo-L-thyronine, T3) on swim bladder inflation and thyroid hormone-related gene expression in Japanese medaka and zebrafish. The swim bladder of most larvae had inflated at 4 h post hatching (hph) in Japanese medaka and at 48 hph in zebrafish in controls. In both fish species, the swim bladder inflation was inhibited in larvae exposed to PFBA (lowest observed effect concentration [LOEC] in medaka: 40 mg/L; in zebrafish: 80 mg/L), TDCPP (LOEC in medaka: 1 mg/L; in zebrafish: 0.5 mg/L), and T3 (no inhibition in Japanese medaka; LOEC in zebrafish: 7.5 µg/L). We also examined the influence of PFBA, TDCPP, and T3 on the expression of thyroid stimulating hormone subunit beta (tshß) or thyroid hormone receptor alpha (trα) and beta (trß). No changes were observed in the expression of genes after PFBA and TDCPP exposure; however, T3 exposure upregulated trα and trß expression in both fish species. When the results were compared between Japanese medaka and zebrafish, swim bladder inflation in both species was found to be inhibited by exposure to thyroid hormone-disrupting chemicals. Our results show that inhibition of the swim bladder inflation at 4 hph in Japanese medaka and 48 hph in zebrafish is a potential indicator of thyroid hormone-disturbing activity of chemicals.


Asunto(s)
Oryzias , Contaminantes Químicos del Agua , Animales , Embrión no Mamífero , Expresión Génica , Larva/metabolismo , Oryzias/genética , Oryzias/metabolismo , Hormonas Tiroideas/metabolismo , Vejiga Urinaria/metabolismo , Contaminantes Químicos del Agua/metabolismo , Contaminantes Químicos del Agua/toxicidad , Pez Cebra/genética , Pez Cebra/metabolismo
14.
Gene ; 822: 146349, 2022 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-35182677

RESUMEN

TDCPP is a flame retardant which has nervous and reproductive toxicity. Although there is a close association between nervous and reproductive system, the exact toxic mechanism of TDCPP in these systems is still seldom, especially in a genome scale. In this study, we explored the transcriptomic landscape of TDCPP in PC12 and GC2 cells using RNAseq method. A total of 465 co-differential expressed genes were found. These genes were mainly enriched in extra-cellular matrix, cell adhesion, cell cycle arrest, oxidoreductase activity GO terms, and PI3K/AKT, focal adhesion, ECM-receptor interaction KEGG pathways. Hub genes (ANXA1, COL27A1, GAS6, GNB4 and THBS1) were extracted using STRING and confirmed by qPCR experiment. Vimentin, HSPA5 and Caspase3 were proved to be responsible to TDCPP in GC2 and PC12 cells. Knockdown assay in PC12 cells showed that these hub genes could also affect the protein expression of vimentin, HSPA5 and Caspase3. In summary, TDCPP might exert its toxic effect through disturbing focal adhesion, ECM-receptor interaction and PI3K/Akt pathways. One of the mechanisms could be influence on the cytoskeleton (vimentin), ER stress (HSPA5) and apoptosis (Caspase3). The sequence data in this study might be a useful resource for future TDCPP related researches.


Asunto(s)
Retardadores de Llama/toxicidad , Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes/efectos de los fármacos , Compuestos Organofosforados/toxicidad , Animales , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Modelos Biológicos , Células PC12 , RNA-Seq , Ratas
15.
Cells ; 11(2)2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-35053312

RESUMEN

Tris(1,3-Dichloro-2-propyl)phosphate (TDCPP) is an organophosphorus flame retardant (OPFR) widely used in a variety of consumer products (plastics, furniture, paints, foams, and electronics). Scientific evidence has affirmed the toxicological effects of TDCPP in in vitro and in vivo test models; however, its genotoxicity and carcinogenic effects in human cells are still obscure. Herein, we present genotoxic and carcinogenic properties of TDCPP in human liver cells (HepG2). 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) and neutral red uptake (NRU) assays demonstrated survival reduction in HepG2 cells after 3 days of exposure at higher concentrations (100-400 µM) of TDCPP. Comet assay and flow cytometric cell cycle experiments showed DNA damage and apoptosis in HepG2 cells after 3 days of TDCPP exposure. TDCPP treatment incremented the intracellular reactive oxygen species (ROS), nitric oxide (NO), Ca2+ influx, and esterase level in exposed cells. HepG2 mitochondrial membrane potential (ΔΨm) significantly declined and cytoplasmic localization of P53, caspase 3, and caspase 9 increased after TDCPP exposure. qPCR array quantification of the human cancer pathway revealed the upregulation of 11 genes and downregulation of two genes in TDCPP-exposed HepG2 cells. Overall, this is the first study to explicitly validate the fact that TDCPP bears the genotoxic, hepatotoxic, and carcinogenic potential, which may jeopardize human health.


Asunto(s)
Carcinógenos/toxicidad , Retardadores de Llama/toxicidad , Hígado/patología , Mutágenos/toxicidad , Compuestos Organofosforados/toxicidad , Calcio/metabolismo , Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Daño del ADN , Esterasas/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células Hep G2 , Humanos , Hígado/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos
16.
J Hazard Mater ; 402: 123784, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33254794

RESUMEN

Tris(1,3-dichloro-2-propyl) phosphate (TDCPP), a widely used chlorinated organophosphorus flame retardant, is an increasingly widespread contaminant of aquatic environment. In this study, time-dependent effect of TDCPP on the freshwater green-algae Chlorella pyrenoidosa was investigated and its underlying mechanisms were explored. We show that TDCPP lower than 10 ppm caused a reversible inhibition of algal growth, with complete inhibition occurring at 15 ppm. This inhibition was not caused by damage from reactive oxygen species, but rather resulted from the impairment of photosynthetic function, with PSII reaction center as the primary target, as indicated by Chl a fluorescence induction, QA- reoxidation, S-state distribution and immunoblot analysis. The reversal of damage caused by TDCPP concentrations under 10 ppm might be attributable to the repair of photosynthetic function by de novo protein biosynthesis in the chloroplast, with the most likely explanation being the replacement of the damaged PSII D1 protein. The results provide novel insights into mechanisms of TDCPP toxicity toward freshwater microalgae and better understanding of ecological consequences of TDCPP in the environment.


Asunto(s)
Chlorella , Retardadores de Llama , Organofosfatos , Compuestos Organofosforados , Fosfatos
17.
Environ Health ; 19(1): 97, 2020 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-32917231

RESUMEN

BACKGROUND: Organophosphate esters (OPEs)-used as flame retardants and plasticizers-are associated with adverse pregnancy outcomes such as reduced fecundity and live births and increased preterm delivery. OPEs may interfere with growth and metabolism via endocrine-disruption, but few studies have investigated endocrine-related outcomes. The objective of this pilot study (n = 56 mother-infant pairs) was to evaluate associations of OPEs with gestational weight gain (GWG), gestational age at delivery, infant anthropometry, and infant feeding behaviors. METHODS: We quantified OPE metabolites (bis-2-chloroethyl phosphate [BCEP], bis (1,3-dichloro-2-propyl) phosphate [BDCPP], diphenyl phosphate [DPHP]) in pooled maternal spot urine collected throughout pregnancy (~ 12, 28, and 35 weeks' gestation). We obtained maternal sociodemographic characteristics from questionnaires administered at enrollment and perinatal characteristics from medical record abstraction. Trained research assistants measured infant weight, length, head and abdominal circumferences, and skinfold thicknesses at birth and 6 weeks postpartum. Mothers reported infant feeding behavior via the Baby Eating Behavior Questionnaire (BEBQ). Using multiple linear regression, we assessed associations of log2-transformed maternal urinary OPE metabolites with GWG, gestational age at delivery, infant anthropometry at birth, weekly growth rate, and BEBQ scores at 6 weeks postpartum. We used linear mixed effects (LME) models to analyze overall infant anthropometry during the first 6 weeks of life. Additionally, we considered effect modification by infant sex. RESULTS: We observed weak positive associations between all OPE metabolites and GWG. In LME models, BDCPP was associated with increased infant length (ß = 0.44 cm, 95%CI = 0.01, 0.87) and weight in males (ß = 0.14 kg, 95%CI = 0.03, 0.24). BDCPP was also associated with increased food responsiveness (ß = 0.23, 95%CI = 0.06, 0.40). DPHP was inversely associated with infant abdominal circumference (ß = - 0.50 cm, 95%CI = - 0.86, - 0.14) and female weight (ß = - 0.19 kg, 95%CI = - 0.36, - 0.02), but positively associated with weekly growth in iliac skinfold thickness (ß = 0.10 mm/wk., 95%CI = 0.02, 0.19). Further, DPHP was weakly associated with increased feeding speed. BCEP was associated with greater infant thigh skinfold thickness (ß = 0.34 mm, 95%CI = 0.16, 0.52) and subscapular skinfold thickness in males (ß = 0.14 mm, 95%CI = 0.002, 0.28). CONCLUSIONS: Collectively, these findings suggest that select OPEs may affect infant anthropometry and feeding behavior, with the most compelling evidence for BDCPP and DPHP.


Asunto(s)
Antropometría , Contaminantes Ambientales/orina , Edad Gestacional , Ganancia de Peso Gestacional/efectos de los fármacos , Recién Nacido/fisiología , Exposición Materna , Organofosfatos/orina , Adulto , Composición Corporal/efectos de los fármacos , Tamaño Corporal/efectos de los fármacos , Ésteres/orina , Conducta Alimentaria/efectos de los fármacos , Femenino , Humanos , Lactante , Recién Nacido/crecimiento & desarrollo , Embarazo , Rhode Island , Adulto Joven
18.
Chemosphere ; 256: 127066, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32434091

RESUMEN

Tri(1,3-dichloropropyl) phosphate (TDCPP) potentially damages the thyroid system in humans and animals. However, knowledge of its toxic effects and underlying mechanisms is limited. The present study was conducted to determine the thyroid hormone-disrupting effects of TDCPP and its major metabolite, bis(1,3-dichloro-2-propyl) phosphate (BDCPP) in rat pituitary cell lines (GH3). TDCPP and BDCPP, that mimic the thyroid hormone (TH), promoted GH3 cell proliferation and modulated the progression of the cell cycle at 20 and 200 µmol/L, respectively. Similar to T3, TDCPP and BDCPP also significantly upregulated c-fos and downregulated Tshß gene expression. Although the binding affinity of these chemicals for thyroid receptor ß (TRß) was not measured, significant competition between these chemicals to bind to the membrane thyroid hormone receptor (integrin αvß3) was found, suggesting that TDCPP and BDCPP were strongly bound to integrin αvß3. Results from a molecular docking analysis provided further evidence of strong binding affinities of TDCPP and BDCPP for integrin αvß3, and the ligand binding site of Arg-Gly-Asp (RGD) was identified. Real-time PCR also supported the supposition that, after binding to integrin αvß3, TDCPP and BDCPP may induce the activation of the extracellular signal-regulated protein kinase (ERK1/2) signal transduction pathway. Taken together, our data suggest that TDCPP and BDCPP have the ability to mimic THs and that the underlying mechanism might be associated with their interactions with integrin αvß3 and the activation of the ERK1/2 pathway, providing new insight into the mechanism of TDCPP- and BDCPP-induced cytotoxicity.


Asunto(s)
Organofosfatos/toxicidad , Hormonas Tiroideas/metabolismo , Pruebas de Toxicidad , Animales , Proliferación Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Integrina alfaVbeta3/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Quinasa 3 Activada por Mitógenos , Simulación del Acoplamiento Molecular , Oligopéptidos , Organofosfatos/metabolismo , Compuestos Organofosforados , Ratas , Transducción de Señal , Glándula Tiroides/efectos de los fármacos
19.
Sci Total Environ ; 726: 138526, 2020 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-32304943

RESUMEN

Tris(1,3-dichloro-2-propyl) phosphate (TDCPP), a widely used organophosphorus flame retardant, has been frequently detected in the environment including indoor dust. Long-term exposure to TDCPP-containing dust may adversely affect human skin, however, little is known about its potential cytotoxicity. In this study, human skin keratinocytes (HaCaT) were employed to study TDCPP-induced cytotoxicity and associated mechanisms. The effects of TDCPP on cell morphology, viability, apoptosis, and cycle, and the mRNA levels of apoptosis (Bcl-2, Bax and Caspase-3) and cell cycle (cyclin D1, CDK2, CDK4 and CDK6) regulatory genes were investigated. The results showed that TDCPP caused a concentration-dependent decrease in cell viability after exposing to TDCPP ≥100 µg/mL for 48 h, with a median lethal concentration of 163 µg/mL (LC50). In addition, TDCPP induced cell apoptosis and arrested cell cycle in the G0/G1 phase at 16 and 160 µg/mL by enhancing Bax and Caspase-3 expression besides inhibiting cyclin D1, CDK2, CDK6 and Bcl-2 expression. Our results showed that TDCPP-induced toxicity in HaCaT cells was probably through cell apoptosis and cell cycle arrest. This study provides information on the toxicity of TDCPP to human skin cells, which may help to reduce its toxicity to human skin.


Asunto(s)
Retardadores de Llama , Apoptosis , Polvo , Humanos , Queratinocitos/química , Organofosfatos , Compuestos Organofosforados/análisis
20.
Arch Toxicol ; 94(2): 541-552, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31894355

RESUMEN

Tris(1,3-dichloro-2-propyl) phosphate (TDCPP) is a phosphorus-based flame retardant common in consumer goods and baby products. Concerns have been raised about TDCPP exposure and neurodevelopmental toxicity. However, the mechanism and early response for TDCPP-induced neurotoxicity are poorly understood. This study investigates the role of microglia-mediated neuroinflammation in TDCPP-induced neurotoxicity in mice and primary cells. TDCPP was administered to C57BL/6 pups (0, 5, or 50 mg/kg/day) via an oral gavage from postnatal days 10-38 (28 days). The results showed that TDCPP exposure for 28 days altered the gene expression of neuronal markers Tubb3, Nefh, and Nes, and led to apoptosis in the hippocampus. The mRNA levels of pro-inflammatory factors Il-1ß, Tnfα and Ccl2 dose dependently increased in the hippocampus at both 24 h and 28 days following exposure, accompanied by microglia activation characterized by an amoeboid-like phenotype. In in vitro studies using the primary microglia isolated from neonatal mice, exposure to TDCPP (0-100 µM) for 24 h resulted in cellular activation. It also increased the expression of genes responsible for inflammatory responses including surface markers and pro-inflammatory cytokines. These changes occurred in a dose-dependent fashion. Neurite outgrowth of primary mouse hippocampal neurons was inhibited by treatment with the conditioned medium harvested from microglia exposed to TDCPP. These results reveal that neonatal exposure to TDCPP induces neuronal damage through microglia-mediated inflammation. This provides insight into the mechanism of TDCPP's neurodevelopmental toxicity, and suggests that microglial cell is a sensitive responder for OPFRs exposure.


Asunto(s)
Retardadores de Llama/toxicidad , Hipocampo/efectos de los fármacos , Microglía/efectos de los fármacos , Síndromes de Neurotoxicidad/patología , Compuestos Organofosforados/toxicidad , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Peso Corporal/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/patología , Hipocampo/fisiología , Inflamación/inducido químicamente , Masculino , Ratones Endogámicos C57BL , Microglía/patología , Síndromes de Neurotoxicidad/etiología , Pruebas de Toxicidad/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA