Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 26
1.
Toxics ; 12(5)2024 May 17.
Article En | MEDLINE | ID: mdl-38787149

Nanoplastics, created by the fragmentation of larger plastic debris, are a serious pollutant posing substantial environmental and health risks. Here, we developed a polystyrene nanoparticle (PS-NP) exposure model during mice pregnancy to explore their effects on embryonic development. We found that exposure to 30 nm PS-NPs during pregnancy resulted in reduced mice placental weight and abnormal embryonic development. Subsequently, our transcriptomic dissection unveiled differential expression in 102 genes under PS-NP exposure and the p38 MAPK pathway emerged as being significantly altered in KEGG pathway mapping. Our findings also included a reduction in the thickness of the trophoblastic layer in the placenta, diminished cell invasion capabilities, and an over-abundance of immature red cells in the blood vessels of the mice. In addition, we validated our findings through the human trophoblastic cell line, HTR-8/SVneo (HTR). PS-NPs induced a drop in the vitality and migration capacities of HTR cells and suppressed the p38 MAPK signaling pathway. This research highlights the embryotoxic effects of nanoplastics on mice, while the verification results from the HTR cells suggest that there could also be certain impacts on the human trophoblast layer, indicating a need for further exploration in this area.

2.
Lipids Health Dis ; 23(1): 126, 2024 Apr 29.
Article En | MEDLINE | ID: mdl-38685082

BACKGROUND: Overweight and obesity are among the leading chronic diseases worldwide. Environmental phenols have been renowned as endocrine disruptors that contribute to weight changes; however, the effects of exposure to mixed phenols on obesity are not well established. METHODS: Using data from adults in National Health and Nutrition Examination Survey, this study examined the individual and combined effects of four phenols on obesity. A combination of traditional logistic regression and two mixed models (weighted quantile sum (WQS) regression and Bayesian kernel-machine regression (BKMR)) were used together to assess the role of phenols in the development of obesity. The potential mediation of cholesterol on these effects was analyzed through a parallel mediation model. RESULTS: The results demonstrated that solitary phenols except triclosan were inversely associated with obesity (P-value < 0.05). The WQS index was also negatively correlated with general obesity (ß: 0.770, 95% CI: 0.644-0.919, P-value = 0.004) and abdominal obesity (ß: 0.781, 95% CI: 0.658-0.928, P-value = 0.004). Consistently, the BKMR model demonstrated the significant joint negative effects of phenols on obesity. The parallel mediation analysis revealed that high-density lipoprotein mediated the effects of all four single phenols on obesity, whereas low-density lipoprotein only mediated the association between benzophenol-3 and obesity. Moreover, Cholesterol acts as a mediator of the association between mixed phenols and obesity. Exposure to single and mixed phenols significantly and negatively correlated with obesity. Cholesterol mediated the association of single and mixed environmental phenols with obesity. CONCLUSIONS: Assessing the potential public health risks of mixed phenols helps to incorporate this information into practical health advice and guidance.


Isoflavones , Obesity , Phenols , Humans , Phenols/urine , Male , Adult , Female , Middle Aged , Cholesterol/blood , Benzhydryl Compounds/urine , Triclosan/adverse effects , Nutrition Surveys , Bayes Theorem , Endocrine Disruptors/urine , Chlorophenols/urine
3.
Front Immunol ; 15: 1372215, 2024.
Article En | MEDLINE | ID: mdl-38655266

Introduction: Lung adenocarcinoma (LUAD) is a prevalent form of lung cancer originating from lung glandular cells with low survival rates despite recent therapeutic advances due to its diverse and complex nature. Recent evidence suggests a link between ferroptosis and the effectiveness of anti-PD-L1 therapy, with potential synergistic effects. Methods: Our study comprehensively analyzed the expression patterns of ferroptosis regulators in LUAD and their association with prognosis and PD-L1 expression. Furthermore, we identified two distinct subtypes of LUAD through consensus clustering of ferroptosis regulators, revealing significant tumor heterogeneity, divergent PD-L1 expression, and varying prognoses between the subtypes. Results: Among the selected ferroptosis regulators, SLC7A11 emerged as an independent prognostic marker for LUAD patients and exhibited a negative correlation with PD-L1 expression. Subsequent investigations revealed high expression of SLC7A11 in the LUAD population. In vitro experiments demonstrated that overexpression of SLC7A11 led to reduced PD-L1 expression and inhibited ferroptosis in A549 cells, underscoring the significant role of SLC7A11 in LUAD. Additionally, pan-cancer analyses indicated an association between SLC7A11 and the expression of immune checkpoint genes across multiple cancer types with poor prognoses. Discussion: From a clinical standpoint, these findings offer a foundation for identifying and optimizing potential combination strategies to enhance the therapeutic effectiveness of immune checkpoint inhibitors and improve the prognosis of patients with LUAD.


Adenocarcinoma of Lung , Amino Acid Transport System y+ , B7-H1 Antigen , Ferroptosis , Gene Expression Regulation, Neoplastic , Lung Neoplasms , Ferroptosis/genetics , Humans , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Amino Acid Transport System y+/genetics , Amino Acid Transport System y+/metabolism , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Prognosis , Down-Regulation , A549 Cells , Biomarkers, Tumor/genetics , Male , Female , Cell Line, Tumor
4.
Ecotoxicol Environ Saf ; 266: 115605, 2023 Nov 01.
Article En | MEDLINE | ID: mdl-37864966

2-Ethylhexyl diphenyl phosphate (EHDPP) is one of the typical organophosphate flame retardants (OPFRs) and has been widely detected in environmental media. Exposure to EHDPP during pregnancy affects placental development and fetal growth. Liver X receptor α (LXRα) is essential to placental development. However, finite information is available regarding the function of LXRα in placenta damages caused by EHDPP. In present study we investigated to figure out whether LXRα is playing roles in EHDPP-induced placenta toxicity. While EHDPP restrained cell viability, migration, and angiogenesis dose-dependently in HTR-8/SVneo and JEG-3 cells, overexpression or activation by agonist T0901317 of LXRα alleviated the above phenomenon, knockdown or inhibition by antagonist GSK2033 had the opposite effects in vitro. Further study indicated EHDPP decreased LXRα expression and transcriptional activity leading to mRNA, protein expression levels downregulation of viability, migration, angiogenesis-related genes Forkhead box M1 (Foxm1), endothelial nitric oxide synthase (eNos), matrix metalloproteinase-2 (Mmp-2), matrix metalloproteinase-9 (Mmp-9), vascular endothelial growth factor-A (Vegf-a) and upregulation of inflammatory genes interleukin-6 (Il-6), interleukin-1ß (Il-1ß) and tumor necrosis factor-α (Tnf-α) in vitro and in vivo. Moreover, EHDPP caused decreased placental volume and fetal weight in mice, treatment with LXRα agonist T0901317 restored these adverse effects. Taken together, our study unveiled EHDPP-induced placenta toxicity and the protective role of LXRα in combating EHDPP-induced placental dysfunction. Activating LXRα could serve as a therapeutic strategy to reverse EHDPP-induced placental toxicity.


Matrix Metalloproteinase 2 , Phosphates , Female , Pregnancy , Animals , Mice , Liver X Receptors/genetics , Vascular Endothelial Growth Factor A , Cell Line, Tumor , Placenta
5.
Environ Pollut ; 293: 118542, 2022 Jan 15.
Article En | MEDLINE | ID: mdl-34801623

Perfluorooctane sulfonic acid (PFOS) is a persistent environmental pollutant. Exposure to PFOS has been associated with abnormal fetal development. The long non-coding RNA (lncRNA) has been showed to play a role in fetal growth restriction (FGR), preeclampsia (PE) and other pregnancy complications. Whether the lncRNA contributes to PFOS-induced toxicity in the placenta remains unknown. In this study, we investigated the function of lncRNA MEG3 and its derived miR-770 in PFOS-induced placental toxicity. Pregnant mice received gavage administration of different concentrations of PFOS (0.5, 2.5, and 12.5 mg/kg/day) from GD0 to GD17, and HTR-8/SVneo cells were treated with PFOS in the concentrations of 0, 10-1, 1, 10 µM. We found that expression levels of miR-770 and its host gene MEG3 were reduced in mice placentas and HTR-8/SVneo cells with exposure of PFOS. A significant hypermethylation was observed at MEG3 promoter in placentas of mice gestational-treated with PFOS. We also confirmed that MEG3 and miR-770 overexpression alleviated the cell growth inhibition induced by PFOS. Furthermore, PTX3 (Pentraxin 3) was identified as the direct target of miR-770 and it was enhanced after PFOS exposure. In summary, our results suggested that MEG3 alleviate PFOS-induced placental cell inhibition through MEG3/miR-770/PTX3 axis.


Alkanesulfonic Acids , Fluorocarbons , MicroRNAs , RNA, Long Noncoding , Alkanesulfonic Acids/toxicity , Animals , C-Reactive Protein , Female , Fluorocarbons/toxicity , Mice , MicroRNAs/genetics , Nerve Tissue Proteins , Placenta , Pregnancy , RNA, Long Noncoding/genetics
6.
iScience ; 24(12): 103487, 2021 Dec 17.
Article En | MEDLINE | ID: mdl-34934913

The loss of uterine epithelial progesterone receptor (PGR) is crucial for successful embryo implantation in both humans and mice. The two major isoforms PGRA and PGRB have divergent functions under both physiological and pathological conditions. The present study compares phenotypes and gene signatures of PGRA and PGRB in uterine epithelium using uterine epithelial-specific constitutively expressed PGRA or PGRB mouse models. The cistrome and transcriptome analysis reveals substantial overlap between epithelial PGRA and PGRB, and both disrupt embryo implantation through FOXO1 pathways. Constitutive epithelial PGRA and PGRB expression impairs ESR1 occupancy at the promoter of Lif leading to reduced Lif transcription and further exaggerates SGK1 expression leading to enhanced PI3K-SGK1 activities, and both contribute to the decline of nuclear FOXO1 expression. Our study demonstrates that PGRA and PGRB in the uterine epithelium act on a similar set of target genes and commonly regulate the LIF-SGK1-FOXO1 signaling pathway for embryo implantation.

7.
Reprod Toxicol ; 106: 18-24, 2021 12.
Article En | MEDLINE | ID: mdl-34547414

2,2',4,4'-Tetrabromodiphenyl ether (BDE47) poses potential risks to reproduction and development, but the mechanism of its toxicity has not yet been elucidated. To explore the developmental toxicity of BDE47, mouse embryonic stem cells (mESCs), which are ideal models for testing the developmental toxicity of environmental contaminants in vitro, were exposed to BDE47 (0.04 µM, 1 µM, 25 µM, or 100 µM) for 24 h or 48 h in this study. Our results indicated that BDE47 treatment changed the morphology of mESCs, inhibited cell viability and increased apoptosis. In addition, alkaline phosphatase (AP) staining in mESCs was significantly decreased after BDE47 treatment (25 µM and 100 µM), indicating that BDE47 treatment affected the pluripotency of mESCs. Through a cell immunofluorescence assay, we found that the fluorescence intensities of Oct4, Sox2 and Nanog were all significantly lower in the group treated with the highest BDE47 concentration (100 µM) than in the control group, consistent with the qRT-PCR and Western blot results. The levels of miR-145 and miR-34a, which regulate genes related to cell differentiation, were significantly increased in BDE47-treated mESCs, further clarifying the potential mechanism. Overall, our findings demonstrate that BDE47 exposure upregulates the expression of miR-145 and miR-34a and in turn downregulates the expression of Oct4, Sox2 and Nanog, thereby affecting apoptosis and pluripotency and causing toxicity during embryonic development.


Halogenated Diphenyl Ethers/toxicity , Mouse Embryonic Stem Cells/drug effects , Alkaline Phosphatase/analysis , Animals , Apoptosis/drug effects , Cells, Cultured , Mice , Mouse Embryonic Stem Cells/enzymology , Mouse Embryonic Stem Cells/physiology
8.
Biomedicines ; 9(6)2021 Jun 15.
Article En | MEDLINE | ID: mdl-34203907

Perfluorooctane sulfonic acid (PFOS), a persistent environmental pollutant, has adverse effects on gestation pregnancy. Peroxisome proliferator-activated receptor γ (PPARγ) is involved in angiogenesis, metabolic processes, anti-inflammatory, and reproductive development. However, the function of PPARγ in PFOS evoked disadvantageous effects on the placenta remain uncertain. Here, we explored the role of PPARγ in PFOS-induced placental toxicity. Cell viability, cell migration, angiogenesis, and mRNA expression were monitored by CCK-8 assay, wound healing assay, tube formation assay, and real-time PCR, respectively. Activation and overexpression of PPARγ were conducted by rosiglitazone or pcDNA-PPARγ, and inhibition and knockdown of PPARγ were performed by GW9662 or si-PPARγ. Results revealed that PFOS decreased cell growth, migration, angiogenesis, and increased inflammation in human HTR-8/SVneo and JEG-3 cells. Placenta diameter and fetal weight decreased in mice treated with PFOS (12.5 mg/kg). In addition, rosiglitazone or pcDNA-PPARγ rescued cell proliferation, migration, angiogenesis, and decreased inflammation induced by PFOS in HTR8/SVneo and JEG-3 cells. Furthermore, GW9662 or si-PPARγ exacerbated the inhibition of cell viability, migration, angiogenesis, and aggravated inflammation induced by PFOS in HTR-8/SVneo and JEG-3 cells. Meanwhile, the results of mRNA expression level were consistent with the cell representation. In conclusion, our findings revealed that PFOS induced placenta cell toxicity and functional damage through PPARγ pathway.

9.
Mol Ther Nucleic Acids ; 24: 104-112, 2021 Jun 04.
Article En | MEDLINE | ID: mdl-33738142

Unexplained recurrent pregnancy loss (URPL) is a significant reproductive health issue, affecting approximately 5% of pregnancies. Enhancer RNAs (eRNAs) have been reported to play important roles during embryo development and may be related to URPL. To investigate whether and how eRNAs are involved in URPL, we performed RNA sequencing in decidual tissue. Through comprehensive screening and validation, we identified a decidua-enriched eRNA long noncoding-CES1-1 (lnc-CES1-1) enriched in URPL patients and studied its function in decidua-associated cell lines (DACs). Higher expression of lnc-CES1-1 increased the level of inflammatory factors tumor necrosis factor alpha (TNF-α) and interleukin-1ß (IL-1ß) and impaired the cell migration ability, which was attenuated by downregulating peroxisome proliferators-activated receptor γ (PPARγ). Upon activation by signal transduction and activation of transcription 4 (STAT4), lnc-CES1-1 interacted with the transcription factor fused in sarcoma (FUS) to upregulate the expression of PPARγ and affected cell migration. Taken together, these findings provide novel insights into the biological functions of decidua-associated lnc-CES1-1 and the molecular mechanisms underlying URPL. Our findings indicated that lnc-CES1-1 might be a potential candidate biomarker for URPL diagnosis and treatment.

10.
Environ Sci Pollut Res Int ; 28(5): 5833-5844, 2021 Feb.
Article En | MEDLINE | ID: mdl-32978737

Heavy metal in the physical environment may alter immune function and predispose to develop asthma in human. Our study was aimed to investigate associations between urinary heavy metals and asthma in adults. A retrospective cross-sectional study was conducted with 3425 subjects aged 20 years and older in the US National Health and Nutrition Examination Survey (NHANES) 2011-2014. Binary logistic regression was applied to analyze associations between cobalt (Co), tungsten (W), and uranium (U) and asthma. We found positive associations between U and asthma (OR = 1.74, 95%CI: 1.25, 2.44, P for trend < 0.01). U was positively associated with asthma in 20-59 years group (OR = 1.65, 95%CI: 1.11, 2.46), while W and Co were related with asthma among in above 60 years group (OR = 2.39, 95%CI: 1.24, 4.58, P for trend = 0.02; OR = 1.88, 95%CI: 1.02, 3.47, respectively). U was linked with asthma in both males and females (OR = 1.93, 95%CI: 1.16, 3.20; OR = 1.59, 95%CI: 1.01, 2.51, respectively). Positive associations between U and asthma were discovered among adults with family history of asthma or not (OR = 2.15, 95%CI: 1.17, 3.95, P for trend = 0.03; OR = 1.62, 95%CI: 1.08, 2.43, P for trend = 0.03, respectively). Remarkable association was observed between U and asthma in adults without hay fever (OR = 1.79, 95%CI: 1.24, 2.60, P for trend = 0.02). Our findings provide epidemiological evidence to highlight a need to prioritize heavy metals exposure with asthma.


Asthma , Metals, Heavy , Adult , Asthma/chemically induced , Asthma/epidemiology , Cross-Sectional Studies , Female , Humans , Logistic Models , Male , Nutrition Surveys , Retrospective Studies , United States/epidemiology , Young Adult
11.
Cells ; 11(1)2021 12 28.
Article En | MEDLINE | ID: mdl-35011648

Exposure to the antibacterial agent triclosan (TCS) is associated with abnormal placenta growth and fetal development during pregnancy. Peroxisome proliferator-activated receptor γ (PPARγ) is crucial in placenta development. However, the mechanism of PPARγ in placenta injury induced by TCS remains unknown. Herein, we demonstrated that PPARγ worked as a protector against TCS-induced toxicity. TCS inhibited cell viability, migration, and angiogenesis dose-dependently in HTR-8/SVneo and JEG-3 cells. Furthermore, TCS downregulated expression of PPARγ and its downstream viability, migration, angiogenesis-related genes HMOX1, ANGPTL4, VEGFA, MMP-2, MMP-9, and upregulated inflammatory genes p65, IL-6, IL-1ß, and TNF-α in vitro and in vivo. Further investigation showed that overexpression or activation (rosiglitazone) alleviated cell viability, migration, angiogenesis inhibition, and inflammatory response caused by TCS, while knockdown or inhibition (GW9662) of PPARγ had the opposite effect. Moreover, TCS caused placenta dysfunction characterized by the significant decrease in weight and size of the placenta and fetus, while PPARγ agonist rosiglitazone alleviated this damage in mice. Taken together, our results illustrated that TCS-induced placenta dysfunction, which was mediated by the PPARγ pathway. Our findings reveal that activation of PPARγ might be a promising strategy against the adverse effects of TCS exposure on the placenta and fetus.


PPAR gamma/metabolism , Placenta/physiopathology , Triclosan/toxicity , Animals , Cell Line , Cell Movement/drug effects , Cell Movement/genetics , Cell Survival/drug effects , Female , Gene Expression Regulation/drug effects , Humans , Inflammation/genetics , Inflammation/pathology , Mice , Models, Biological , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , Placenta/drug effects , Pregnancy
12.
J Cell Mol Med ; 24(12): 7072-7077, 2020 06.
Article En | MEDLINE | ID: mdl-32349189

Recurrent pregnancy loss (RPL) rates have continued to rise during the last few decades, yet the underlying mechanisms remain poorly understood. An emerging area of interest is the mediation of gene expression by DNA methylation during early pregnancy. Here, genome-wide DNA methylation from placental villi was profiled in both RPL patients and controls. Subsequently, differentially expressed genes were analysed for changes in gene expression. Many significant differentially methylated regions (DMRs) were identified near genes dysregulated in RPL including PRDM1. Differentially expressed genes were enriched in immune response pathways indicating that abnormal immune regulation contributes to RPL. Integrated analysis of DNA methylome and transcriptome demonstrated that the expression level of PRDM1 is fine-tuned by DNA methylation. Specifically, hypomethylation near the transcription start site of PRDM1 can recruit other transcription factors, like FOXA1 and GATA2, leading to up-regulation of gene expression and resulting in changes to trophoblast cell apoptosis and migration. These phenotypic differences may be involved in RPL. Overall, our study provides new insights into PRDM1-dependent regulatory effects during RPL and suggests both a mechanistic link between changes in PRDM1 expression, as well as a role for PRDM1 methylation as a potential biomarker for RPL diagnosis.


Abortion, Habitual/genetics , DNA Methylation/genetics , Positive Regulatory Domain I-Binding Factor 1/genetics , Apoptosis/genetics , Case-Control Studies , Cell Cycle/genetics , Cell Movement/genetics , Female , GATA2 Transcription Factor/metabolism , Gene Expression Regulation , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Pregnancy , Trophoblasts/metabolism
13.
J Hazard Mater ; 385: 121534, 2020 03 05.
Article En | MEDLINE | ID: mdl-31706747

Di-(2-ethylhexyl)-phthalate (DEHP) is a ubiquitous environmental pollutant and is widely used in industrial plastics. However, the long-term health implications of prenatal exposure to DEHP remains unclear. We set out to determine whether prenatal DEHP exposure can induce metabolic syndrome in offspring and investigate the underlying mechanisms. A mouse model of prenatal DEHP exposure (0.2, 2, and 20 mg/kg/day) was established to evaluate the long-term metabolic disturbance in offspring. The mice were profiled for the hepatic metabolome, transcriptome and gut microbiota to determine the underlying mechanisms. Thiamine supplementation (50 mg/kg/day) was administered to offspring to investigate the role of thiamine in ameliorating metabolic syndrome. Prenatal exposure to low-dose DEHP (0.2 mg/kg/day) resulted in metabolic syndrome, including abnormal adipogenesis, energy expenditure and glucose metabolism, along with dysbiosis of the gut microbiome, in male offspring. Notably, hepatic thiamine metabolism was disrupted in these offspring due to the dysregulation of thiamine transport enzymes, which caused abnormal glucose metabolism. Prenatal low-dose DEHP exposure caused life-long metabolic consequences in a sex-dependent manner, and these consequences were be attenuated by thiamine supplementation in offspring. Our findings suggest low-dose DEHP exposure during early life stages is a potential risk factor for later obesity and metabolic syndrome.


Diethylhexyl Phthalate/toxicity , Environmental Pollutants/toxicity , Metabolic Syndrome/chemically induced , Obesity/chemically induced , Prenatal Exposure Delayed Effects/chemically induced , Thiamine/pharmacology , Animals , Dysbiosis/chemically induced , Dysbiosis/metabolism , Female , Gastrointestinal Microbiome/drug effects , Gene Expression/genetics , Liver/drug effects , Male , Maternal Exposure/adverse effects , Metabolic Syndrome/metabolism , Metabolome/drug effects , Mice, Inbred ICR , Obesity/metabolism , Pregnancy , Risk Factors , Transcriptome/drug effects
14.
Mol Med Rep ; 20(4): 3802-3810, 2019 Oct.
Article En | MEDLINE | ID: mdl-31485625

Glial cell line­derived neurotrophic factor (GDNF) is critical for the proliferation of spermatogonial stem cells (SSCs), but the underlying mechanisms remain poorly understood. In this study, an unbiased metabolomic analysis was performed to examine the metabolic modifications in SSCs following GDNF deprivation, and 11 metabolites were observed to decrease while three increased. Of the 11 decreased metabolites identified, glycylglycine was observed to significantly rescue the proliferation of the impaired SSCs, while no such effect was observed by adding sorbitol. However, the expression of self­renewal genes, including B­cell CLL/lymphoma 6 member B, ETS variant 5, GDNF family receptor α1 and early growth response protein 4 remained unaltered following glycylglycine treatment. This finding suggests that although glycylglycine serves an important role in the proliferation of SSCs, it is not required for the self­renewal of SSCs.


Glycylglycine/metabolism , Spermatogonia/cytology , Stem Cells/cytology , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Male , Metabolome , Mice, Inbred C57BL , Spermatogonia/metabolism , Stem Cells/metabolism
15.
Mol Ther Nucleic Acids ; 16: 605-615, 2019 Jun 07.
Article En | MEDLINE | ID: mdl-31085354

Long noncoding RNAs (lncRNAs) have been reported to be involved in various human diseases, and increasing studies have revealed that lncRNAs can play a vital role in preeclampsia (PE). In our study, lncRNA hypoxia-inducible factor 1 alpha (HIF1A) antisense RNA 2 (HIF1A-AS2) was found to be significantly downregulated in placenta tissues of PE patients by quantitative real-time PCR analysis. Moreover, Cell Counting Kit-8 (CCK-8) assays showed that downregulation of HIF1A-AS2 can impede cell proliferation of HTR-8/SVneo and JAR trophoblasts cells. Ectopic overexpression of HIF1A-AS2 can increase the function of trophoblasts cell migration and invasion in vitro. RNA-sequencing (RNA-seq), RNA immunoprecipitation (RIP), and chromatin immunoprecipitation (ChIP) experiments showed that HIF1A-AS2 can recruit lysine-specific demethylase 1 (LSD1) and epigenetically repress pleckstrin homology-like domain, family A, member 1 (PHLDA1) transcription in human trophoblasts cells. In summary, our findings suggest that downregulated HIF1A-AS2 may play a role in the pathogenesis and progression of PE, and has potential as a novel prognostic marker in PE.

16.
Nanotoxicology ; 13(2): 204-220, 2019 03.
Article En | MEDLINE | ID: mdl-30663479

Iron oxides nanoparticles (FeOX NPs), including α-Fe2O3, γ-Fe2O3, and Fe3O4, are employed in many technological applications. However, very few studies have investigated the embryonic developmental toxicity of FeOX NPs. In this study, metabolomics analysis were used to uncover the potential mechanisms of FeOX NPs developmental toxicity on embryo-larval zebrafish and mice. Our results indicated that γ-Fe2O3 NP treatment could cause increased mortality, dropped hatching rate, etc., while α-Fe2O3 and Fe3O4 NPs showed no obvious effect. Through metabolomics analysis, a total of 42 metabolites were found to be significantly changed between the γ-Fe2O3 NP-treated group and the control group (p < 0.05). Pathway enrichment analysis indicated the impairment of mitochondria function. γ-Fe2O3 NP treatment caused abnormal mitochondrion structure and a decrease in mitochondrial membrane potential in zebrafish embryos. Meanwhile, ATP synthesis was decreased while oxidative stress levels were affected. It is noteworthy that acetyl-l-carnitine (ALCAR) (p = 6.79E - 04) and l-carnitine (p = 1.43E - 03) were identified with minimal p values, the relationship between the two counter-balance was regulated by acetyltransferase (crata). Subsequently, we performed rescue experiments with ALCAR on zebrafish embryos, and found that the mortality rates reduced and hatching rates raised significantly in the γ-Fe2O3 NP-treated group. Additionally, γ-Fe2O3 exposure could lead to increased absorbed fetus rate, decreased placental weight, lower expression of acetyltransferase (Crat), reduced ATP synthesis as well as increased oxidative stress (p < 0.05). Our findings demonstrated that γ-Fe2O3 NP might affect the mitochondrial membrane potential and ATP synthesis by affecting the metabolism of ALCAR, thereby stimulating oxidative stress, cell apoptosis, and causing embryonic development toxicity.


Acetylcarnitine/metabolism , Embryo, Nonmammalian/drug effects , Embryonic Development/drug effects , Ferric Compounds/toxicity , Mitochondria/drug effects , Nanoparticles/toxicity , Zebrafish , Animals , Embryo, Nonmammalian/metabolism , Female , Ferric Compounds/chemistry , Membrane Potential, Mitochondrial/drug effects , Metabolomics , Mice , Nanoparticles/chemistry , Oxidative Stress/drug effects , Pregnancy
17.
Ecotoxicol Environ Saf ; 169: 829-836, 2019 Mar.
Article En | MEDLINE | ID: mdl-30597782

Pentachlorophenol (PCP) is often used as chlorinated hydrocarbon herbicides and insecticides, which has been suggested that toxicity of carcinogenic effect, teratogenic effect and reproductive system. However, there was still precious known about the underlying molecular mechanism of PCP on mammalian early development. To explore the developmental toxicity of PCP and its potential mechanism, pregnancy ICR mice except controls were exposed to PCP (0.02, 0.2 or 2 mg/kg) during gestation day (GD) 0.5 to GD8.5 in this study. We found that the fetal loss rate was increased and placental chorionic villi structure was disorder in hematoxylin-eosin staining (HE) on GD16.5. Meanwhile, autophagosomes were observed in chorionic villi through Transmission Electron Microscope (TEM). Moreover, the mRNA and/or protein expression of P62, LC3-ІІ/LC3-І and Beclin1 were increased in placenta, indicating the occurrence of autophagy. Then, to further explore the autophagy mechanism, microRNA (miR)-30a-5p, an expression inhibitor of Beclin1, was predicted through bioinformatics predictions and RT-PCR, and it was reduced in PCP-treated mice. Transfection and luciferase reporter gene test were used to verify the interaction between Beclin1 and miR-30a-5p. These results firstly indicate that, PCP exposure could downregulate the expression of miR-30a-5p, and then induced autophagy through upregulation of Beclin1 to result in fetal loss. Our study laid a foundation for understanding the PCP developmental toxicity through autophagy.


Abortion, Spontaneous/chemically induced , Autophagy/drug effects , Environmental Pollutants/toxicity , Maternal Exposure/adverse effects , Pentachlorophenol/toxicity , Abortion, Spontaneous/genetics , Abortion, Spontaneous/pathology , Animals , Beclin-1/genetics , Disease Models, Animal , Down-Regulation , Female , Humans , Mice, Inbred ICR , MicroRNAs/genetics , Placenta/drug effects , Placenta/pathology , Pregnancy , Up-Regulation
18.
Ecotoxicol Environ Saf ; 167: 412-421, 2019 Jan 15.
Article En | MEDLINE | ID: mdl-30368134

Perfluorooctanoate (PFOA) and perfluorooctane sulfonate (PFOS) are widespread and persistent chemicals in the environment, and limited data about their effects on puberty development are available. In order to explore the effects of neonatal and juvenile PFOA/PFOS exposure on puberty maturation, female rats were injected with PFOA or PFOS at 0.1, 1 and 10 mg/kg/day during postnatal day (PND) 1-5 or 26-30. The day of vaginal opening (VO) and first estrus were significantly advanced in 10 mg/kg PFOA, 1 and 10 mg/kg PFOS groups after neonatal and juvenile exposure. Besides, neonatal PFOA/PFOS exposure increased body weight and anogenital distance (AGD) in a non-dose-dependent manner. Estradiol and luteinizing hormone levels were also increased with more frequent occurrences of irregular estrous cycles in 0.1 and 1 mg/kg PFOA/PFOS exposure groups. Although no altered ovarian morphology was observed, follicles numbers were reduced in neonatal groups. Kiss1, Kiss1r and ERα mRNA expressions were downregulated after two periods' exposure in the hypothalamic anteroventral periventricular (AVPV) and arcuate (ARC) nuclei. PFOA/PFOS exposure also suppressed kisspeptin fiber intensities, especially at the high dose. In conclusion, neonatal and juvenile are critical exposure periods, during which puberty maturation may be vulnerable to environmental exposure of PFOA/PFOS, and kisspeptin system plays a key role during these processes.


Alkanesulfonic Acids/toxicity , Caprylates/toxicity , Fluorocarbons/toxicity , Hypothalamus/metabolism , Kisspeptins/metabolism , Sexual Maturation/drug effects , Animals , Body Weight/drug effects , Down-Regulation/drug effects , Estradiol/blood , Estrogen Receptor alpha/genetics , Estrous Cycle/drug effects , Female , Kisspeptins/genetics , Luteinizing Hormone/blood , Ovarian Follicle/drug effects , RNA, Messenger/metabolism , Rats , Receptors, Kisspeptin-1/genetics
19.
EBioMedicine ; 38: 162-170, 2018 Dec.
Article En | MEDLINE | ID: mdl-30448228

BACKGROUND: Enhancer RNAs (eRNAs) are a group of lncRNAs transcribed from enhancers, whose regulatory effects on gene expression are an emerging area of interest. However, the role of eRNAs in regulating trophoblast cells and unexplained recurrent pregnancy loss (URPL) remains elusive. METHODS: We profiled eRNAs in villi from URPL patients and matched controls by RNA-seq. Functions of URPL-related eRNAs were further investigated in vitro. RESULTS: We identified lnc-SLC4A1-1, which was transcribed from an active enhancer marked with H3K27ac and H3K4me1 and so-called eRNA, highly expressed in URPL patients. Gain-of-function experiments indicated that lnc-SLC4A1-1 facilitated trophoblast cell migration and apoptosis. Mechanistically, as an eRNA, lnc-SLC4A1-1 was retained in the nuclei and recruited transcription factor NF-κB to bind to CXCL8, resulting in increased H3K27ac in the CXCL8 promoter and subsequent elevation of CXCL8 expression. Activation of CXCL8 exacerbated inflammatory reactions in trophoblast cells by inducing TNF-α and IL-1ß, which could be blocked by an antagonist of lnc-SLC4A1-1. INTERPRETATION: These findings indicate that an eRNA, lnc-SLC4A1-1, alters trophoblast function via activation of immune responses and by regulating the NF-κB/CXCL8 axis. Our study provides new insights in understanding lncRNA/eRNA function in pathological pregnancy, potentially informing on therapeutic strategies for URPL. FUND: National Natural Science Foundation of China, Natural Science Foundation of Jiangsu Province, National Key Research and Development Program, the Priority Academic Program for the Development of Jiangsu Higher Education Institutions.


Abortion, Habitual/genetics , Abortion, Habitual/metabolism , Anion Exchange Protein 1, Erythrocyte/genetics , Epigenesis, Genetic , Interleukin-8/metabolism , NF-kappa B/metabolism , RNA, Long Noncoding/genetics , Abortion, Habitual/immunology , Case-Control Studies , Computational Biology/methods , Disease Susceptibility , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Models, Biological , Pregnancy , Protein Binding , Signal Transduction
20.
EBioMedicine ; 35: 334-344, 2018 Sep.
Article En | MEDLINE | ID: mdl-30100398

BACKGROUND: Aberrant DNA methylation is considered to be a potential cause of recurrent pregnancy loss (RPL), while potential mechanism has not yet been elucidated. METHODS: In order to uncover the contribution of the perturbation of DNA methylation in RPL, we performed genome-wide DNA methylation analysis combined with genome-wide gene expression in decidua tissue. FINDINGS: Totally, 539 differentially methylated regions (DMRs) were identified and significantly correlated with gene expressions. We observed that hypo-methylated DMR near CREB5 recruited transcription factors binding, such as P53 and SP1, and in turn upregulated CREB5. Compromised cell migration and apoptosis were observed in human CREB5 overexpression trophoblast cell lines, indicating dysfunctional trophoblast cells might contribute to RPL after hypo-methylation of CREB5. In addition, overexpression of CREB5 altered cell cycle. INTERPRETATION: Our data highlights a role of CREB5 involved in the pathogenesis of RPL, and CREB5 maybe a potential diagnostic biomarker for RPL.


Abortion, Habitual/genetics , Cyclic AMP Response Element-Binding Protein A/genetics , DNA Methylation/genetics , Genetic Predisposition to Disease , Transcriptome/genetics , Adult , Base Sequence , Case-Control Studies , Cell Line , Cyclic AMP Response Element-Binding Protein A/metabolism , Decidua/metabolism , Female , Gene Expression Regulation , Gene Knockdown Techniques , Genome, Human , Humans , Pregnancy , Risk Factors , Sp1 Transcription Factor/metabolism , Tumor Suppressor Protein p53/metabolism , Young Adult
...