Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Environ Sci Technol ; 58(15): 6475-6486, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38578163

ABSTRACT

Arsenic (As) is widely present in the natural environment, and exposure to it can lead to learning and memory impairment. However, the underlying epigenetic mechanisms are still largely unclear. This study aimed to reveal the role of histone modifications in environmental levels of arsenic (sodium arsenite) exposure-induced learning and memory dysfunction in male rats, and the inter/transgenerational effects of paternal arsenic exposure were also investigated. It was found that arsenic exposure impaired the learning and memory ability of F0 rats and down-regulated the expression of cognition-related genes Bdnf, c-Fos, mGlur1, Nmdar1, and Gria2 in the hippocampus. We also observed that inorganic arsenite was methylated to DMA and histone modification-related metabolites were altered, contributing to the dysregulation of H3K4me1/2/3, H3K9me1/2/3, and H3K4ac in rat hippocampus after exposure. Therefore, it is suggested that arsenic methylation and hippocampal metabolism changes attenuated H3K4me1/2/3 and H3K4ac while enhancing H3K9me1/2/3, which repressed the key gene expressions, leading to cognitive impairment in rats exposed to arsenic. In addition, paternal arsenic exposure induced transgenerational effects of learning and memory disorder in F2 male rats through the regulation of H3K4me2 and H3K9me1/2/3, which inhibited c-Fos, mGlur1, and Nmdar1 expression. These results provide novel insights into the molecular mechanism of arsenic-induced neurotoxicity and highlight the risk of neurological deficits in offspring with paternal exposure to arsenic.


Subject(s)
Arsenic , Rats , Animals , Male , Arsenic/toxicity , Histone Code , Hippocampus , Methylation
2.
J Appl Toxicol ; 43(2): 262-271, 2023 02.
Article in English | MEDLINE | ID: mdl-35978532

ABSTRACT

Ubiquitous micro(nano)plastics (MNPs) are emerging environmental pollutants, which pose a potential threat to human health. When MNPs enter the blood circulatory system, vascular endothelium is one of the most important target organs that directly interact with the MNPs. However, little is known about the cytotoxicity of MNPs to vascular endothelial cells. In this study, we investigated the uptake and cytotoxic effects of polystyrene MNPs with a particle size of 1 µm (1-µm PS-MNPs) on human umbilical vein endothelial cells (HUVECs) in vitro. Our study found that interaction between HUVECs and 1-µm PS-MNPs was at a very low level. Even at the high exposure concentration of 25 µg/mL, the percentage of HUVECs combined with fluorescent 1-µm PS-MNPs was only 3.80% using flow cytometry analysis. Moreover, there were no significant differences in inflammation, autophagy, reactive oxygen species (ROS) level, lactate dehydrogenase (LDH) release, and adhesion molecule expression following exposure to 1-µm PS-MNPs (5, 10, and 25 µg/mL) for 48 h, except for a remarkable decrease in cell viability at the extremely high concentration of 100 µg/mL. Herein, 1-µm PS-MNPs showed a low level of acute toxicity to HUVECs in vitro, and we expect these results contribute to the further risk assessment of MNPs on human health.


Subject(s)
Microplastics , Water Pollutants, Chemical , Humans , Human Umbilical Vein Endothelial Cells/metabolism , Microplastics/toxicity , Polystyrenes/toxicity , Plastics/metabolism , Reactive Oxygen Species/metabolism , Water Pollutants, Chemical/metabolism
3.
Environ Sci Technol ; 56(9): 5664-5672, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35438966

ABSTRACT

Perfluorooctanoic acid (PFOA) is a persistent organic pollutant, which has endocrine-disrupting properties and can interfere with the synthesis and secretion of testicular steroid hormones, but the underlying molecular mechanisms are still not fully understood. In this study, we investigated the effects of low doses of PFOA exposure on testicular steroidogenesis in rats and revealed the role of histone modifications. It was found that the serum levels of progesterone, testosterone, and estradiol were significantly increased after 0.015 and 0.15 mg/kg of PFOA exposure, and the expression of Star, a key rate-limiting gene, was up-regulated, while other steroidogenic genes Cyp11a1, Hsd3b, Cyp17a1, and Hsd17b were down-regulated. In addition, the levels of multiple histone modifications (H3K9me1/2/3 and H3K9/18/23ac) were all significantly reduced by PFOA in rat testis. Histone H3K9 methylation is associated with gene silencing, while histone acetylation leads to gene activation. ChIP analysis further showed that H3K9me1/3 was significantly decreased in the promoter region of Star, while H3K18ac levels were down-regulated in other gene promoters. Accordingly, we suggest that low-level PFOA enhances StAR expression through the repression of H3K9me1/3, which stimulates steroid hormone production in rat testis. These results are expected to shed new light on the molecular mechanisms by which low-dose PFOA disturbs male reproductive endocrine from an epigenetic aspect and may be useful for human health risk assessment regarding environmental PFOA exposure.


Subject(s)
Histones , Testosterone , Animals , Caprylates , Fluorocarbons , Histones/metabolism , Male , Methylation , Rats , Steroids , Testosterone/metabolism
4.
J Acoust Soc Am ; 146(6): 4097, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31893714

ABSTRACT

Based on the distribution of acoustic intensity at different depths at a fixed distance, a simple method is proposed to estimate the sound source depth at known range in the deep sea. First, the method calculates the acoustic intensity distribution of all the depths at a receiving distance. Second, the depths with the strongest acoustic intensity are selected. Sound sources are set at the selected depths to calculate the transmission loss (TL) at the same distance through the acoustic model, and the depth of the minimum superimposed TL is considered as the depth of the original sound source. The simulation and experiment verify the feasibility and reliability of the method.

5.
Arch Toxicol ; 92(3): 1023-1035, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29222745

ABSTRACT

Exposure to ambient particulate matter (PM) has been linked to the increasing incidence and mortality of lung cancer, but the principal toxic components and molecular mechanism remain to be further elucidated. In this study, human lung adenocarcinoma A549 cells were treated with serial concentrations of water-extracted PM10 (WE-PM10) collected from Beijing, China. Our results showed that exposure to 25 and 50 µg/ml of WE-PM10 for 48 h significantly suppressed miR-26a to upregulate lin-28 homolog B (LIN28B), and in turn activated interleukin 6 (IL6) and signal transducer and activator of transcription 3 (STAT3) in A549 cells, subsequently contributing to enhanced epithelial-mesenchymal transition and accelerated migration and invasion. In vivo pulmonary colonization assay further indicated that WE-PM10 enhanced the metastatic ability of A549 cells. In addition, luciferase reporter assay demonstrated that 3' untranslated region of LIN28B was a direct target of miR-26a. Last but not the least, the key toxic contribution of metals in WE-PM10 was confirmed by the finding that removal of metals through chelation significantly rescued WE-PM10-mediated inflammatory, carcinogenic and metastatic responses. Taken together, miR-26a could act as the tumor suppressor in PM10-related lung cancer, and PM10-bound metals promoted lung cancer cell metastasis through downregulation of miR-26a that directly mediated LIN28B expression.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , MicroRNAs/genetics , Particulate Matter/toxicity , RNA-Binding Proteins/genetics , A549 Cells , Animals , Cell Movement/drug effects , Cell Movement/genetics , Humans , Inflammation/chemically induced , Inflammation/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Metals/analysis , Metals/toxicity , Mice, Inbred BALB C , Particulate Matter/chemistry , RNA-Binding Proteins/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Xenograft Model Antitumor Assays
6.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(9): 869-882, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28483554

ABSTRACT

Exposure to Bisphenol A (BPA) has been associated with the development of nonalcoholic fatty liver disease (NAFLD) but the underlying mechanism remains unclear. Given that microRNA (miRNA) is recognized as a key regulator of lipid metabolism and a potential mediator of environmental cues, this study was designed to explore whether exposure to BPA-triggered abnormal steatosis and lipid accumulation in the liver could be modulated by miR-192. We showed that male post-weaning C57BL/6 mice exposed to 50µg/kg/day of BPA by oral gavage for 90days displayed a NAFLD-like phenotype. In addition, we found in mouse liver and human HepG2 cells that BPA-induced hepatic steatosis and lipid accumulation were associated with decreased expression of miR-192, upregulation of SREBF1 and a series of genes involved in de novo lipogenesis. Downregulation of miR-192 in BPA-exposed hepatocytes could be due to defective pre-miR-192 processing by DROSHA. Using HepG2 cells, we further confirmed that miR-192 directly acted on the 3'UTR of SREBF1, contributing to dysregulation of lipid homeostasis in hepatocytes. MiR-192 mimic and lentivirus-mediated overexpression of miR-192 improved BPA-induced hepatic steatosis by suppressing SREBF1. Lastly, we noted that lipid accumulation was not a strict requirement for developing insulin resistance in mice after BPA treatment. In conclusion, this study demonstrated a novel mechanism in which NAFLD associated with BPA exposure arose from alterations in the miR-192-SREBF1 axis.


Subject(s)
Benzhydryl Compounds/pharmacology , Down-Regulation/genetics , Fatty Liver/pathology , Lipids/physiology , MicroRNAs/genetics , Non-alcoholic Fatty Liver Disease/genetics , Phenols/pharmacology , Sterol Regulatory Element Binding Protein 1/genetics , Animals , Cell Line, Tumor , Fatty Liver/genetics , Fatty Liver/metabolism , Hep G2 Cells , Humans , Lipid Metabolism/genetics , Lipogenesis/drug effects , Lipogenesis/genetics , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Sterol Regulatory Element Binding Protein 1/metabolism , Up-Regulation/genetics
7.
Environ Pollut ; 350: 124030, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38663511

ABSTRACT

As a widely used alternative to perfluorooctanoic acid (PFOA), hexafluoropropylene oxide trimer acid (HFPO-TA) has been detected in the environment and humans; however, little is known regarding its male reproductive toxicity. To compare the effects of HFPO-TA on steroid hormone synthesis with PFOA, we exposed Leydig cells (MLTC-1) to non-lethal doses (0.1, 1, and 10 µM) of PFOA and HFPO-TA for 48 h. It was found that the levels of steroid hormones, 17α-hydroxyprogesterone (OHP), androstenedione (ASD), and testosterone (T) were significantly increased in 1 and 10 µM of PFOA and HFPO-TA groups, with greater elevation being observed in the HFPO-TA groups than in the PFOA groups at 10 µM. We further showed that the two rate-limiting steroidogenic genes (Star and Cyp11a1) were up-regulated, while Hsd3b, Cyp17a1, and Hsd17b were down-regulated or unchanged after PFOA/HFPO-TA exposure. Moreover, PFOA exposure significantly up-regulated histone H3K4me1/3 and H3K9me1, while down-regulated H3K4me2 and H3K9me2/3 levels. By contrast, H3K4me2/3 and H3K9me2/3 were enhanced, while H3K4me1 and H3K9me1 were repressed after HFPO-TA treatment. It was further confirmed that H3K4me1/3 were increased and H3K9me2 was decreased in Star and Cyp11a1 promoters by PFOA, while HFPO-TA increased H3K4me2/3 and decreased H3K9me1 in the two gene promoters. Therefore, we propose that low levels of PFOA/HFPO-TA enhance the expression of Star and Cyp11a1 by regulating H3K4 and H3K9 methylation, thus stimulating the production of steroid hormones in MLTC-1 cells. Collectively, HFPO-TA exhibits stronger effects on steroidogenesis compared to PFOA, which may be ascribed to the distinct regulation of histone modifications. These data suggest that HFPO-TA does not appear to be a safer alternative to PFOA on the aspect of male reproductive toxicity.


Subject(s)
Caprylates , Fluorocarbons , Fluorocarbons/toxicity , Caprylates/toxicity , Animals , Male , Histone Code/drug effects , Leydig Cells/drug effects , Leydig Cells/metabolism , Testosterone/metabolism , Histones/metabolism , Mice
8.
J Hazard Mater ; 468: 133796, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38377905

ABSTRACT

Haloacetic acids (HAAs) are ubiquitous in drinking water and have been associated with impaired male reproductive health. However, epidemiological evidence exploring the associations between HAA exposure and reproductive hormones among males is scarce. In the current study, the urinary concentrations of dichloroacetic acid (DCAA) and trichloroacetic acid (TCAA), the internal exposure markers of HAAs, as well as sex hormones (testosterone [T], progesterone [P], and estradiol [E2]) were measured among 449 Chinese men. Moreover, in vitro experiments, designed to simulate the real-world scenarios of human exposure, were conducted to assess testosterone synthesis in the Leydig cell line MLTC-1 and testosterone metabolism in the hepatic cell line HepG2 in response to low-dose HAA exposure. The DCAA and TCAA urinary concentrations were found to be positively associated with urinary T, P, and E2 levels (all p < 0.001), but negatively associated with the ratio of urinary T to E2 (p < 0.05). Combined with in vitro experiments, the results suggest that environmentally-relevant doses of HAA stimulate sex hormone synthesis and steroidogenesis pathway gene expression in MLTC-1 cells. In addition, the inhibition of the key gene CYP3A4 involved in the testosterone phase Ⅰ catabolism, and induction of the gene UGT2B15 involved in testosterone phase Ⅱ glucuronide conjugation metabolism along with the ATP-binding cassette (ABC) transport genes (ABCC4 and ABCG2) in HepG2 cells could play a role in elevation of urinary hormone excretion upon low-dose exposure to HAAs. Our novel findings highlight that exposure to HAAs at environmentally-relevant concentrations is associated with increased synthesis and excretion of sex hormones in males, which potentially provides an alternative approach involving urinary hormones for the noninvasive evaluation of male reproductive health following exposure to DBPs.


Subject(s)
Disinfection , Drinking Water , Humans , Male , Trichloroacetic Acid/toxicity , Dichloroacetic Acid/analysis , Dichloroacetic Acid/urine , Steroids , Testosterone
9.
Environ Int ; 185: 108532, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38422876

ABSTRACT

Nanoplastics (NPs) continue to accumulate in global aquatic and terrestrial systems, posing a potential threat to human health through the food chain and/or other pathways. Both in vivo and in vitro studies have confirmed that the liver is one of the main organs targeted for the accumulation of NPs in living organisms. However, whether exposure to NPs induces size-dependent disorders of liver lipid metabolism remains controversial, and the reversibility of NPs-induced hepatotoxicity is largely unknown. In this study, the effects of long-term exposure to environmentally relevant doses of polystyrene nanoplastics (PS-NPs) on lipid accumulation were investigated in terms of autophagy and lysosomal mechanisms. The findings indicated that hepatic lipid accumulation was more pronounced in mice exposed to 100 nm PS-NPs compared to 500 nm PS-NPs. This effect was effectively alleviated after 50 days of self-recovery for 100 nm and 500 nm PS-NPs exposure. Mechanistically, although PS-NPs exposure activated autophagosome formation through ERK (mitogen-activated protein kinase 1)/mTOR (mechanistic target of rapamycin kinase) signaling pathway, the inhibition of Rab7 (RAB7, member RAS oncogene family), CTSB (cathepsin B), and CTSD (cathepsin D) expression impaired lysosomal function, thereby blocking autophagic flux and contributing to hepatic lipid accumulation. After termination of PS-NPs exposure, lysosomal exocytosis was responsible for the clearance of PS-NPs accumulated in lysosomes. Furthermore, impaired lysosomal function and autophagic flux inhibition were effectively alleviated. This might be the main reason for the alleviation of PS-NPs-induced lipid accumulation after recovery. Collectively, we demonstrate for the first time that lysosomes play a dual role in the persistence and reversibility of hepatotoxicity induced by environmental relevant doses of NPs, which provide novel evidence for the prevention and intervention of liver injury associated with nanoplastics exposure.


Subject(s)
Chemical and Drug Induced Liver Injury , Nanoparticles , Water Pollutants, Chemical , Humans , Animals , Mice , Microplastics , Polystyrenes/toxicity , Lysosomes , Lipids
10.
ISA Trans ; 137: 175-185, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36639267

ABSTRACT

This paper is concerned with the measurement outlier-resistant mobile robot localization problem by using multiple Doppler-azimuth radars under round-robin protocol (R-RP). In the considered robot localization system, multiple Doppler-azimuth radars are equipped on the robot platform to produce the measurement including the Doppler frequency shift and the azimuth. In order to assuage communication link congestion, the R-RP is used. For mitigating the influence of outliers, a time-varying state estimator is constructed which contains a saturation function with variable saturation levels. This paper aims at seeking out a practicable yet effective solution to the addressed robot localization problem by devising the constructed estimator which can assure that, over a finite horizon, the localization error satisfies the given H∞ performance index. By constructing an appropriate Lyapunov function, the sufficient condition, which can guarantee the localization error to fulfill the given H∞ performance, is established. Then, by resorting to the solution to a set of linear matrix inequalities, the constructed estimator can be devised. In the light of the estimator design strategy proposed in this paper, the corresponding robot localization algorithm is developed. At last, some simulations are conducted to testify the usefulness of the developed robot localization algorithm.

11.
Genes Genomics ; 45(11): 1423-1431, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37646913

ABSTRACT

BACKGROUND: Endometrial carcinoma (EC) is the most prevalent gynecological cancer. Transcription factor (TF) regulates a large number of downstream target genes and is a key determinant of all physiological activities, including cell proliferation, differentiation, apoptosis, and cell cycle. The transcription factor E2F1 shows prominent roles in EC. BMI1 is a member of Polycomb suppressor Complex 1 (PRC1) and has been shown to be associated with EC invasiveness. It is currently unclear whether E2F1 can participate in the proliferation, migration, and invasion processes of EC cells by regulating BMI1 transcription. OBJECTIVE: We investigated whether E2F1 could participate in the proliferation, migration, and invasion processes of EC cells by regulating BMI1 transcription, in order to further clarify the pathogenesis and etiology of EC, and provide reference for identifying potential therapeutic targets and developing effective prevention and treatment strategies for this disease. METHODS: Human endometrial epithelial cells (hEECs) and human EC cell lines were selected. E2F1 expression was assessed by Western blot. E2F1 was silenced in AN3CA or overexpressed in HEC-1 by transfections, or E2F1 was silenced and BMI1 was overexpressed in AN3CA by cotransfection. Cell proliferation, migration, and invasion were detected by MTT, wound healing, and Transwell assays. The binding sites between E2F1 and BMI1 promoters were predicted through JASPAR website, and the targeted binding was verified by dual-luciferase report and ChIP assays. RESULTS: E2F1 was up-regulated in human EC cell lines, with its expression highest in AN3CA, and lowest in HEC-1. AN3CA invasion, migration, and proliferation were repressed by E2F1 knockdown, while those of HEC-1 cells were promoted by E2F1 overexpression. E2F1 overexpression increased the activity of wild type BMI1 reporter vector promoter, while this promotion was weakened after mutation of the predicted binding site in the BMI1 promoter. In the precipitated E2F1, BMI1 promoter site level was higher than that of IgG immunoprecipitant. BMI1 silencing suppressed AN3CA cell growth. BMI1 overexpression partially abrogated E2F1 silencing-inhibited EC cell growth. CONCLUSION: E2F1 promoted EC cell proliferation, invasion, and migration by promoting the transcription of BMI1.

12.
J Hazard Mater ; 445: 130623, 2023 03 05.
Article in English | MEDLINE | ID: mdl-37056006

ABSTRACT

Autophagy was involved in vascular endothelial injury caused by PM2.5, which aggravated the pathogenesis of cardiovascular diseases. However, major toxic components and underlying mechanism responsible for PM2.5-induced autophagy remain unclear. In this study, the effects of water-extracted PM2.5 (WE-PM2.5) on autophagy in human umbilical vein endothelial cells (HUVEC) were studied. Our results showed WE-PM2.5 promoted autophagosome initiation and formation, meanwhile, lysosomal function was impaired, which further caused autophagic flux blockage in HUVEC cells. Furthermore, removal of metals alleviated WE-PM2.5-induced autophagic flux blockage, while the artificial metal mixture reproduced the WE-PM2.5 response. Mechanistically, ROS regulated autophagy-related proteins evidenced by BECN1, LC3B and p62 expression reversed by NAC pretreatment in WE-PM2.5-exposed cells. WE-PM2.5 also increased TXNIP expression mediated by ROS; moreover, knockdown of TXNIP in WE-PM2.5-exposed cells decreased BECN1 and LC3B expression, but had little effects on the expression of p62, CTSB, and CTSD, indicating WE-PM2.5-induced TXNIP was involved in autophagosome initiation and formation rather than autophagic degradation. Collectively, WE-PM2.5-induced ROS not only promoted autophagosome initiation and formation, but also inhibited autophagic degradation. However, as the downstream molecule of ROS, TXNIP was only involved in autophagosome initiation and formation. Importantly, WE-PM2.5-bound metals were largely responsible for autophagic flux blockage in HUVEC cells.


Subject(s)
Autophagosomes , Autophagy , Humans , Human Umbilical Vein Endothelial Cells , Reactive Oxygen Species/metabolism , Autophagosomes/metabolism , Autophagosomes/pathology , Metals/metabolism , Particulate Matter/toxicity , Particulate Matter/metabolism , Carrier Proteins/metabolism
13.
J Reprod Immunol ; 158: 103971, 2023 08.
Article in English | MEDLINE | ID: mdl-37329866

ABSTRACT

OBJECTIVE: Stem cell/exosome therapy is a novel strategy for primary ovarian insufficiency (POI). This paper is to examine the role of human umbilical cord mesenchymal stem cell-derived extracellular vesicles (hUCMSC-EVs) in POI. METHODS: hUCMSC-EVs were extracted and identified. POI rats were induced by cyclophosphamide for 15 days and treated with EV or GW4869 every 5 days and euthanized 28 days later. Vaginal smears were observed for 21 days. Serum hormone levels (FSH/E2/AMH) were measured by ELISA. Ovarian morphology, follicle numbers, and granulosa cell (GC) apoptosis were observed by HE and TUNEL staining. GCs extracted from Swiss albino rats were cyclophosphamide-induced to establish the POI cell model, followed by oxidative injury and apoptosis evaluation with the help of DCF-DA fluorescence, ELISA, and flow cytometry. The relation between miR-145-5p and XBP1 was predicted on StarBase and validated by dual-luciferase assay. miR-145-5p and XBP1 levels were measured by RT-qPCR and Western blot. RESULTS: EV treatment reduced irregular estrus cycle incidence since day 7, increased E2 and AMH levels and all-stage follicle numbers, reduced FSH level, GC apoptosis, and atretic follicle numbers in POI rats. EV treatment diminished GC oxidative injury and apoptosis in vitro. miR-145-5p knockdown in hUCMSC-EVs partly abolished hUCMSC-EV-mediated effects on GCs and ovarian function in vivo and on GC oxidative injury and apoptosis in vitro. Silencing XBP1 partially negated miR-145-5p knockdown-exerted effects on GCs in vitro. CONCLUSION: miR-145-5p carried by hUCMSC-EVs attenuates GC oxidative injury and apoptosis and thus extenuates ovarian injury and improves ovarian function in POI rats.


Subject(s)
Exosomes , Mesenchymal Stem Cells , MicroRNAs , Primary Ovarian Insufficiency , Female , Rats , Humans , Animals , Primary Ovarian Insufficiency/therapy , Cyclophosphamide/adverse effects , Cyclophosphamide/metabolism , Exosomes/metabolism , Umbilical Cord/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Mesenchymal Stem Cells/metabolism , Follicle Stimulating Hormone
14.
Sci Total Environ ; 892: 164761, 2023 Sep 20.
Article in English | MEDLINE | ID: mdl-37315596

ABSTRACT

Environmental arsenic (As) exposure has been associated with gestational diabetes mellitus (GDM) risk. Our recent study found that GDM was positively associated with urinary As3+ level while negatively correlated to As5+. However, the mechanisms underlying the association between arsenic species and GDM remain largely unknown. In the present study, through the measurement of urinary arsenic species and metabolome analysis in 399 pregnant women, we aimed to identify the metabolic biomarkers that may link arsenic exposure to GDM based on a novel systems epidemiology strategy termed meet-in-metabolite-analysis (MIMA). The metabolomics analysis revealed that 20 and 16 urinary metabolites were relevant to arsenic exposure and GDM, respectively. Among them, 12 metabolites were identified to be both arsenic- and GDM-related, which are mainly involved in purine metabolism, one­carbon metabolism (OCM) and glycometabolism. Moreover, it was further showed that the regulation of thiosulfate (AOR: 2.52; 95 % CI: 1.33, 4.77) and phosphoroselenoic acid (AOR: 2.35; 95 % CI: 1.31, 4.22) could significantly contribute to the negative association between As5+ and GDM. Considering the biological functions of these metabolites, it is suggested that As5+ may reduce GDM risk by disturbing OCM in the pregnant women. These data will provide novel insights into the mechanism of action of environmental arsenic exposure on GDM incidence from the aspect of metabolism disorder.


Subject(s)
Arsenic , Diabetes, Gestational , Pregnancy , Humans , Female , Diabetes, Gestational/epidemiology , Arsenic/urine , Pregnant Women , Cross-Sectional Studies , East Asian People , Biomarkers/metabolism
15.
J Hazard Mater ; 436: 129213, 2022 08 15.
Article in English | MEDLINE | ID: mdl-35739735

ABSTRACT

Humans are simultaneously and constantly exposed to various lipophilic chain phthalate acid esters. The association of urinary phthalate metabolites with altered male steroid hormone synthesis and metabolism was examined using epidemiology and toxicology studies. We measured 8 phthalate metabolites [monomethyl phthalate (MMP), monoethyl phthalate (MEP), mono-n-butyl phthalate (MBP), mono-benzyl phthalate (MBzP), mono-n-octylphthalate (MOP), mono-(2-ethylhexyl) phthalate (MEHP), mono-(2-ethyl-5-hydroxyhexyl) phthalate (MEHHP) and mono (2-ethyl-5-oxohexyl) phthalate (MEOHP)] and two sex hormones [testosterone (T) and estradiol (E2)] in single serum and repeated spot urine samples among 451 reproductive-age males. Moreover, in vitro experiments with Leydig cell MLTC-1 steroidogenesis and liver cell HepG2 efflux in response to mixed and individual phthalates were designed to simulate real-world scenarios of human exposure. As a joint mixture, the phthalate metabolite was inversely associated with serum T and E2 concentrations but positively associated with urinary T and E2 concentrations. Combined with in vitro experiments, DEHP metabolites were identified as the predominant contributor to the decline in hormone synthesis, and ATP-binding cassette (ABC) gene activation might be involved in hormone excretion. Exposure to environmentally relevant phthalates was associated with both altered steroid synthesis and excretion, which provides additional insights into the endocrine-disrupting potential of phthalates.


Subject(s)
Environmental Pollutants , Phthalic Acids , Environmental Exposure , Environmental Pollutants/metabolism , Hormones , Humans , Male , Phthalic Acids/metabolism , Reproduction , Steroids
16.
J Hazard Mater ; 421: 126770, 2022 01 05.
Article in English | MEDLINE | ID: mdl-34358975

ABSTRACT

Ubiquitous nanoplastics (NPs) increase exposure risks to humans through the food chain and/or other ways. However, huge knowledge gaps exist regarding the fate and adverse impact of NPs on the human cardiovascular system. Autophagy is an important catabolic pathway that disposes of cytoplasmic waste through the lysosomes. In this study, we pursued to determine the interaction and autophagy effect of polystyrene nanoplastics (PS-NPs) (100 and 500 nm in size) on human umbilical vein endothelial cells (HUVECs). The results showed both sizes of PS-NPs interacted with almost all the treated HUVECs in a time- and concentration-dependent manner, and 500 nm PS-NPs were only bound to the surface of cell membranes, whereas 100 nm PS-NPs were taken up by HUVECs and aggregated in the cytoplasm. Furthermore, exposure to 25 µg/mL of 500 nm PS-NPs for 48 h significantly increased lactate dehydrogenase release from HUVECs, while internalized 100 nm PS-NPs not only caused cell membrane damage, but also induced autophagy initiation and autophagosome formation. By a mCherry-GFP-LC3 lentivirus infection assay, we also demonstrated that autophagic flux level was impaired in response to 100 nm PS-NPs. Herein, our results provide new insight into the size-dependent internalization and autophagy response to PS-NPs in HUVECs.


Subject(s)
Nanoparticles , Polystyrenes , Autophagy , Human Umbilical Vein Endothelial Cells , Humans , Lysosomes , Microplastics , Nanoparticles/toxicity
17.
Toxicology ; 420: 11-20, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30935970

ABSTRACT

Although the production of polychlorinated biphenyl 77 (PCB77) has already been banned globally, PCB77 is still used for a wide range of commercial purposes. Previous evidence has demonstrated that the PCB77 administration should be responsible for the gut microbiota variations and the host health risk. However, the host disorders and bacterial functions involved in PCB77 exposure remain largely unknown. Few studies have been performed to illuminate the correlation between the bacterial functions and disorders. Furthermore, it is urgently needed to find specific strains as potential biomarkers to monitor PCB77 pollution and associated disorders. This study was designed to investigate the effects of PCB77 on gut microbiota and induced disorders in female mice. Obtained results indicated that PCB77 exposure induced gut microbiota dysbiosis, obesity, hyperlipidemia, hepatic lipid accumulation, and liver injury in mice. Functional prediction based on the phylogenetic investigation of communities by reconstruction of unobserved states (PICRUSt) algorithm showed that exposure to PCB77 weakened the bacterial functions relating to lipid and energy metabolism, and immune system disease. Experimental findings were consistent with the result of the PICRUSt functional prediction. Importantly, three PCB77-associated bacterial taxa were screened out as potential biomarkers for the assessment of PCB77 pollution. This study provides previously unknown knowledge linking PCB77 administration, gut microbiota functional profile and lipid abnormalities, which is of important clinical significance for therapies treating PCB77-associated diseases.


Subject(s)
Bacteria/drug effects , Energy Metabolism/drug effects , Fatty Liver/chemically induced , Gastrointestinal Microbiome/drug effects , Hyperlipidemias/chemically induced , Intestines/microbiology , Lipids/blood , Obesity/chemically induced , Polychlorinated Biphenyls/toxicity , Animals , Bacteria/metabolism , Biomarkers/blood , Dysbiosis , Fatty Liver/blood , Fatty Liver/microbiology , Female , Hyperlipidemias/blood , Hyperlipidemias/microbiology , Mice, Inbred C57BL , Obesity/blood , Obesity/microbiology , Risk Factors , Sex Factors
18.
Sci Total Environ ; 653: 274-282, 2019 Feb 25.
Article in English | MEDLINE | ID: mdl-30412872

ABSTRACT

There is growing evidence that polychlorinated biphenyl 126 (PCB126) not only has adverse effects on host health but also has the ability to shift gut microbiota, which is recently recognized as a crucial factor determining numerous physiological processes. However, the interplay between the gut microbiota and host health remains largely unknown. Herein, adult female C57BL/6 mice were orally exposed to environmentally relevant low-dose of PCB126, at 50 µg/kg body weight once per week for 6 weeks. This study aims to illuminate how PCB126 influences gut microbiota variations and host disorders and to further identify the correlation between the gut microbiota and metabolic markers of host disorders. Obtained results demonstrated that the PCB126 administration induced gut microbiota dysbiosis in mice, with changes both in the gut microbiota constitution and structure. PCB126 administration also simultaneously altered the physiological status of serum and liver, as evaluated by dyslipidemia, liver lipid accumulation and injury, and non-alcoholic fatty liver disease. Importantly, Spearman's correlation analysis suggested that several specific bacterial taxa were positively and significantly related to metabolic markers of the mentioned disorders. Moreover, based on the co-occurrence network map, some of the bacterial taxa may synergistically regulate host physiology. This work provides new insight into the mechanism underlying the interaction between the gut microbiota and host disorders. It is expected that gut microbiota modulation should be another novel way used for the prevention and treatment of PCB126-triggered diseases.


Subject(s)
Dysbiosis/chemically induced , Dyslipidemias/chemically induced , Gastrointestinal Microbiome/drug effects , Non-alcoholic Fatty Liver Disease/chemically induced , Polychlorinated Biphenyls/toxicity , Animals , Female , Mice , Mice, Inbred C57BL
19.
Environ Pollut ; 248: 269-278, 2019 May.
Article in English | MEDLINE | ID: mdl-30798028

ABSTRACT

Exposure to ambient particular matters (PM) has been associated with the development of non-alcoholic fatty liver disease (NAFLD), but the underlying mechanism remains unclear. Given that microRNA (miRNA) is recognized as a key regulator of lipid metabolism and a potential mediator of environmental cues, this study aimed to explore the role of miRNA-mRNA regulation underlying abnormal lipid metabolism triggered by PM2.5liposoluble extracts. We confirmed that 72-h exposure to liposoluble extracts of PM2.5 from Nanjing at 25 µg/cm2 induced lipid accumulation in HepG2 cells by promoting uptake of free fatty acids (FFAs). Notably, lipid accumulation induced by PM2.5 liposoluble extracts was associated with decreased expression of miR-26a and consequent upregulation of fatty acid translocase (FAT, also known as CD36). Using gain- and loss-of-function assays, we demonstrated that miR-26a negatively regulated CD36 to mediate lipid accumulation in HepG2 cells. We further confirmed that miR-26a directly acted on the 3' untranslated region (3'UTR) of CD36. Furthermore, overexpression of miR-26a abolished steatosis in HepG2 cells treated with PM2.5 liposoluble extracts by suppressing CD36. In addition, we demonstrated that PM2.5 liposoluble extracts caused inflammation in HepG2 cells by raising p65 phosphorylation, thereby fuelling the transition from simple non-alcoholic fatty liver to non-alcoholic steatohepatitis. In conclusion, this study demonstrated a novel mechanism by which miR-26a-CD36 pathway mediated lipid accumulation induced by PM2.5 liposoluble extracts in hepatocytes. Lipid accumulation and inflammation induced by PM2.5 liposoluble extracts implied the potential role of PM2.5 in developing NAFLD.


Subject(s)
Air Pollutants/toxicity , Particulate Matter/toxicity , Toxicity Tests , Animals , Biological Transport , Hep G2 Cells , Hepatocytes , Humans , Lipid Metabolism , Lipids , Liver/metabolism , MicroRNAs/metabolism , Non-alcoholic Fatty Liver Disease , Particulate Matter/metabolism , Phosphorylation , RNA, Messenger/metabolism , Signal Transduction , Up-Regulation
20.
Sci Rep ; 7(1): 9392, 2017 08 24.
Article in English | MEDLINE | ID: mdl-28839203

ABSTRACT

Fine particulate matter (PM2.5) exposure, especially to its organic components, induces adverse health effects on the respiratory system. However, the molecular mechanisms have still not been fully elucidated. Long non-coding RNA (lncRNA) is involved in various physio-pathological processes. In this study, the roles of lncRNA were investigated to reveal the toxicology of PM2.5. Organic extracts of PM2.5 from Nanjing and Shanghai cities were adopted to treat human bronchial epithelial cell lines (BEAS-2B and A549). RNA sequencing showed that the lncRNA functioned as antisense RNA, intergenic RNA and pre-miRNA. The mRNA profiles were also altered after exposure. PM2.5 from Nanjing showed a more serious impact than that from Shanghai. In detail, higher expression of n405968 was positively related to the elevated mRNA levels of inflammatory factors (IL-6 and IL-8). Increasing levels of metastasis associated lung adenocarcinoma transcript 1 (MALAT1) were positively associated with the induced epithelial-mesenchymal transition (EMT) process. Similar response was observed between both cell lines. The higher content of polycyclic aromatic hydrocarbons (PAHs) is likely to contribute to higher toxicity of PM2.5 from Nanjing than that from Shanghai. Antagonism of aryl hydrocarbon receptor (AHR) or inhibition of CYP1A1 diminished the effects stimulated by PM2.5. Our results indicated that lncRNAs could be involved in the toxicology of PM2.5 through regulating the inflammation and EMT process.


Subject(s)
Gene Expression Regulation/drug effects , Particulate Matter/toxicity , RNA, Long Noncoding , Air Pollutants/adverse effects , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Phenotype , RNA Interference , RNA, Messenger/genetics , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL