Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.803
Filtrar
1.
Mol Reprod Dev ; 91(4): e23742, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38644727

RESUMO

Preeclampsia (PE) is a common pregnancy complication with a high mortality rate. Abnormally activated endoplasmic reticulum stress (ERS) is believed to be responsible for the destruction of key placental cells-trophoblasts. Phenylbutyric acid (4-PBA), an ERS inhibitor, is involved in regulating the development of ERS-related diseases. At present, how 4-PBA affects trophoblasts and its mechanisms is still unclear. In this study, PE cell models were established by stimulating HTR-8/SVneo cells with hypoxia. To verify the underlying mechanisms of 4-PBA on PE, CCT020312, an activator of PERK, was also used. The results showed that 4-PBA restored hypoxia-induced trophoblast viability, inhibited HIF-1α protein expression, inflammation, and PERK/ATF-4/CHOP pathway. Hoechst 33342 staining and flow cytometry results confirmed that 4-PBA decreased hypoxia-induced apoptosis in trophoblasts. The results of the JC-1 analysis and apoptosis initiation enzyme activity assay also demonstrated that 4-PBA inhibited apoptosis related to the mitochondrial pathway. Furthermore, by detecting autophagy in trophoblasts, an increased number of autophagic vesicles, damaged mitochondria, enhanced dansylcadaverine fluorescence, enhanced levels of autophagy proteins Beclin-1, LC3II, and decreased p62 were seen in hypoxia-stimulated cells. These changes were reversed by 4-PBA. Furthermore, it was observed that CCT020312 reversed the effects of 4-PBA on the viability, apoptosis, and autophagosome number of hypoxia-induced trophoblasts. In summary, 4-PBA reduces autophagy and apoptosis via the PERK/ATF-4/CHOP pathway and mitochondrial pathway, thereby restoring the viability of hypoxic trophoblasts. These findings provide a solid evidence base for the use of 4-PBA in PE treatment and guide a new direction for improving the outcomes of patients with PE.


Assuntos
Fator 4 Ativador da Transcrição , Apoptose , Autofagia , Hipóxia Celular , Fenilbutiratos , Pré-Eclâmpsia , Fator de Transcrição CHOP , Trofoblastos , eIF-2 Quinase , Trofoblastos/efeitos dos fármacos , Trofoblastos/metabolismo , Trofoblastos/patologia , Feminino , Humanos , Pré-Eclâmpsia/metabolismo , Pré-Eclâmpsia/tratamento farmacológico , Pré-Eclâmpsia/patologia , Autofagia/efeitos dos fármacos , Fator de Transcrição CHOP/metabolismo , Apoptose/efeitos dos fármacos , Gravidez , Fenilbutiratos/farmacologia , eIF-2 Quinase/metabolismo , Fator 4 Ativador da Transcrição/metabolismo , Hipóxia Celular/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Linhagem Celular
2.
Cells ; 13(2)2024 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-38247805

RESUMO

Low-dose aspirin (LDA) is efficacious in preventing preeclampsia, but its mechanism of action is unclear. Conflicting evidence suggests that it may inhibit placental trophoblast release of soluble fms-like tyrosine kinase-1 (sFlt1), a key mediator of preeclampsia. We examined whether, and at what concentrations, aspirin and its principal metabolite, salicylic acid, modulate sFlt1 release and/or expression in trophoblasts. Human trophoblast lines BeWo and HTR-8/SVneo were cultured; BeWo cells were also treated with 1% oxygen vs. normoxia to mimic hypoxia in preeclamptic placentas. Cells were treated with aspirin or salicylic acid vs. vehicle for 24 h at concentrations relevant to LDA and at higher concentrations. Protein concentrations (ELISA) and mRNA expression (RT-PCR) of sFlt1 were determined. Under normoxia, LDA-relevant concentrations of aspirin (10-50 µmol/L) or salicylic acid (20-100 µmol/L) had no significant effect on sFlt1 protein release or mRNA expression in BeWo cells. However, inhibition was observed at higher concentrations (1 mmol/L for aspirin and ≥200 µmol/L for salicylic acid). Hypoxia enhanced sFlt1 protein release and mRNA expression in BeWo cells, but these responses were not significantly affected by either aspirin or salicylic acid at LDA concentrations. Similarly, neither drug altered sFlt1 protein secretion or mRNA expression in normoxic HTR-8/SVneo cells at LDA concentrations. We suggest that direct modulation of trophoblast release or expression of sFlt1 is unlikely to be a mechanism underlying the clinical efficacy of LDA in preeclampsia.


Assuntos
Aspirina , Pré-Eclâmpsia , Trofoblastos , Receptor 1 de Fatores de Crescimento do Endotélio Vascular , Feminino , Humanos , Gravidez , Aspirina/farmacologia , Hipóxia , Placenta , Pré-Eclâmpsia/tratamento farmacológico , Receptores Proteína Tirosina Quinases , RNA Mensageiro/genética , Ácido Salicílico/farmacologia , Trofoblastos/efeitos dos fármacos , Trofoblastos/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/efeitos dos fármacos
3.
Mol Med Rep ; 25(4)2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35137920

RESUMO

Pentraxin 3 (PTX3), a member of the c­reactive protein family, is a long pentraxin protein and a pro­inflammatory marker. However, the role of PTX3 in preeclampsia (PE) remains to be elucidated. Thus, the present study aimed to investigate the biological role and mechanisms underlying PTX3 in PE. In the present study, PTX3 was overexpressed in trophoblasts and the subsequent changes in cell proliferation, cycle distribution and invasion were observed using Cell Counting Kit­8, flow cytometry and Transwell assays, respectively. Moreover, the expression levels of MMP2 and MMP9, proteins associated with the development of PE, were detected using reverse transcription­quantitative PCR and western blot analysis. Following treatment with interleukin (IL)­1ß, the expression levels of PTX3 were measured. Furthermore, subsequent changes in cell proliferation, cycle distribution and invasion were investigated following overexpression of PTX3 and treatment with IL­1 receptor antagonist (IL­1Ra). Overexpression of PTX3 inhibited the proliferation, cycle and invasion of HTR­8/SV neo and JEG3 cells. Moreover, treatment with IL­1ß increased the expression of PTX3 in HTR­8/SV neo and JEG3 cells, which was suppressed following treatment with the IL­1ß antagonist. Following PTX3 overexpression and treatment with IL­1Ra, the inhibitory effects of PTX3 overexpression alone on the invasion of HTR­8/SV neo and JEG3 cells were attenuated. In conclusion, these results indicated that IL­1ß could induce PTX3 upregulation, which led to the inhibition of the proliferation, invasion and cell cycle of trophoblasts, thereby promoting the progression of PE.


Assuntos
Proteína C-Reativa/metabolismo , Ciclo Celular/genética , Movimento Celular/genética , Proliferação de Células/genética , Interleucina-1beta/farmacologia , Pré-Eclâmpsia/metabolismo , Componente Amiloide P Sérico/metabolismo , Trofoblastos/metabolismo , Proteína C-Reativa/genética , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Interleucina-1beta/antagonistas & inibidores , Pré-Eclâmpsia/genética , Gravidez , Componente Amiloide P Sérico/genética , Trofoblastos/efeitos dos fármacos
4.
Toxicol In Vitro ; 80: 105327, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35134484

RESUMO

Tumor necrosis factor (TNF) regulates trophoblast turnover during the formation of the placental syncytium and can be a potentially relevant target for adverse effects of xenobiotics. We mimicked syncytialization in vitro by stimulating BeWo cells with 50 µM forskolin. Undifferentiated and syncytialized BeWo cells were exposed to TNF (10 pg/mL-10 ng/mL) for 48 h after which cell viability, progesterone release and gene expression of a selected set of markers representative for placental function were assessed. In undifferentiated BeWo cells, high TNF levels (1-10 ng/mL) increased gene expression of TNF, NF-κB, and TNFRSF1B to maximally 99 ± 17, 2.2 ± 0.2, and 3.0 ± 0.4 of control values, respectively (p < 0.001). These effects were also found in syncytialized BeWo cells but less pronounced. Additionally, TNF may induce syncytialization in BeWo cells as it upregulated ERVW-1 expression by 1.55 ± 0.14-fold (p < 0.05). On the contrary, TNF levels of 10 and 100 pg/mL did not affect gene expression in both undifferentiated and syncytialized BeWo cells, but did enhance cell viability in syncytialised BeWo cells (p < 0.001). In conclusion, we found that high TNF levels (1-10 ng/mL) increased gene expression of TNF, NF-κB, and TNFRSF1B especially in undifferentiated BeWo cells, while physiological TNF concentrations positively affected cell viability and while there was no effect on any of the investigated functional markers.


Assuntos
Trofoblastos/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Colforsina/farmacologia , Feminino , Expressão Gênica , Humanos , Gravidez , Progesterona/metabolismo , Trofoblastos/metabolismo
5.
Toxicol In Vitro ; 80: 105328, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35150872

RESUMO

Proper migration and invasion of extravillous trophoblast cells into the endometrium in early gestation is essential for successful embryo implantation. The development of nanotechnology has led to the emergence of nickel nanoparticles (Ni NPs), for which attendant health concerns are widespread. Ni NPs are known to affect reproduction and be embryotoxic, but whether they affect the migration and invasion functions of trophoblast cells is unclear. We investigated the effects of Ni NPs on the migration and invasion of HTR-8/SVneo in extravillous trophoblast cells and explored the possible role of the PI3K/AKT/MMP2 signaling pathway in this regard. Results showed that the migration and invasion of cells was significantly inhibited by the exposure of Ni NPs. The protein and mRNA levels of PI3K/AKT/MMP2 signaling pathway were significantly reduced with the increase in Ni NPs concentration. The presence of the PI3K activator 740Y-P partially attenuated the inhibition of cell migration and invasion by Ni NPs, confirming the involvement of this pathway. Thus, Ni NPs inhibit migration and invasion of human trophoblast HTR-8/SVneo cells by downregulating the PI3K/AKT/MMP2 signaling pathway. This study is important for the development of safety evaluation criteria for Ni NPs.


Assuntos
Nanopartículas Metálicas/toxicidade , Níquel/toxicidade , Trofoblastos/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Humanos , Metaloproteinase 2 da Matriz/genética , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Trofoblastos/metabolismo
6.
Bioengineered ; 13(1): 1174-1184, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34982020

RESUMO

Gestational diabetes mellitus (GDM) is a complication developed during pregnancy and recover after childbirth. The purpose of this study was to investigate the protective role of FOXC1 during GDM and the underlying mechanism. FOXC1 was downregulated in GDM placental tissues and HG-treated HTR-8/SVneo cells. Overexpression of FOXC1 prevented HG-induced inhibition of cell proliferation, migration and invasion. FOXC1 suppressed HG-induced cell apoptosis in HTR-8/SVneo cells. The apoptosis-related proteins: cleaved caspase-3, cleaved caspase-9 and BAX, were also downregulated by FOXC1 overexpression. FOXC1 increased glucose uptake and improved insulin sensitivity. The expression of FOXC1 was positively correlated with FGF19 expression. FOXC1 regulated the expression of FGF19 and phosphorylation of AMPK. Inhibition of FGF19 attenuated the biological functions of FOXC1 through inactivation of AMPK. In conclusion, this study demonstrates that FOXC1 attenuates HG-induced trophoblast cell injury through upregulating FGF19 to activate the AMPK signaling pathway during GDM, suggesting that FOXC1 is a potential therapeutic target for drug discovery in the future.


Assuntos
Adenilato Quinase/metabolismo , Diabetes Gestacional/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Glucose/efeitos adversos , Trofoblastos/citologia , Adulto , Estudos de Casos e Controles , Diabetes Gestacional/induzido quimicamente , Regulação para Baixo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Idade Materna , Fosforilação , Gravidez , Transdução de Sinais , Trofoblastos/efeitos dos fármacos , Trofoblastos/metabolismo
7.
Reprod Toxicol ; 108: 10-17, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34995713

RESUMO

Environmental pollutant dioxins are potentially harmful to pregnant women and can lead to severe adverse outcomes in pregnancy, such as spontaneous abortion and stillbirth. However, little is currently known about the underlying toxicological mechanism. Our previous study reported that the IL-24 gene is a dioxin response gene during 2, 3, 7, 8-tetrachlorodibenzo-p-dioxin (TCDD) treatment. Here, we further tested the effect of TCDD on IL-24 expression in human chorionic stromal cells. We also investigated the effect of IL-24 on the behaviors of human placental trophoblast cells and predicted the potential mechanism underlying these behaviors using functional network analysis. We found that TCDD stimulates IL-24 expression in human chorionic stromal cells in an AhR (aromatic hydrocarbon receptor)-related manner. We also found that IL-24 inhibits the migration and invasion of human placental trophoblast cells, the possible mechanism of which involves thirteen key proteins and mitochondrial function. Our findings suggest that IL-24 is a potential factor induced by TCDD to regulate trophoblast cell invasion, which potentially involves in TCDD-induced abortion.


Assuntos
Poluentes Ambientais/toxicidade , Interleucinas/metabolismo , Dibenzodioxinas Policloradas/toxicidade , Células Estromais/efeitos dos fármacos , Trofoblastos/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Córion/citologia , Citocromo P-450 CYP1A1/genética , Humanos , Interleucinas/genética , Proteoma/efeitos dos fármacos , Células Estromais/metabolismo , Transcriptoma/efeitos dos fármacos , Trofoblastos/fisiologia
8.
Int J Mol Sci ; 23(2)2022 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-35054855

RESUMO

The placenta supports fetal growth and is vulnerable to exogenous chemical exposures. We have previously demonstrated that exposure to the emerging chemical bisphenol S (BPS) can alter placental endocrine function. Mechanistically, we have demonstrated that BPS interferes with epidermal growth factor receptor (EGFR) signaling, reducing placenta cell fusion. Extravillous trophoblasts (EVTs), a placenta cell type that aids with vascular remodeling, require EGF to invade into the maternal endometrium. We hypothesized that BPS would impair EGF-mediated invasion and proliferation in EVTs. Using human EVTs (HTR-8/SVneo cells), we tested whether BPS could inhibit the EGF response by blocking EGFR activation. We also evaluated functional endpoints of EGFR signaling, including EGF endocytosis, cell invasion and proliferation, and endovascular differentiation. We demonstrated that BPS blocked EGF-induced phosphorylation of EGFR by acting as a competitive antagonist to EGFR. Transwell assay and a three-dimensional microfluidic chip invasion assay revealed that BPS exposure can block EGF-mediated cell invasion. BPS also blocked EGF-mediated proliferation and endovascular differentiation. In conclusion, BPS can prevent EGF-mediated EVT proliferation and invasion through EGFR antagonism. Given the role of EGFR in trophoblast proliferation and differentiation during placental development, our findings suggest that maternal exposure to BPS may contribute to placental dysfunction via EGFR-mediated mechanisms.


Assuntos
Receptores ErbB/metabolismo , Fenóis/toxicidade , Transdução de Sinais , Sulfonas/toxicidade , Trofoblastos/patologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Colágeno/farmacologia , Combinação de Medicamentos , Endocitose/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , Humanos , Laminina/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteoglicanas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Trofoblastos/efeitos dos fármacos
9.
Arch Toxicol ; 96(2): 559-570, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35048155

RESUMO

Prothioconazole (PTC) is a new broad-spectrum triazole antibacterial agent that is being widely used in agriculture. PTC has been linked to a number of reproductive outcomes including embryo implantation disorder; however, the exact mechanism underlying this relationship has yet to be determined. Proper trophoblast proliferation and migration is a prerequisite for successful embryo implantation. To elucidate the underlying molecular perturbations, we detect the effect of PTC on extravillous trophoblast cells proliferation and migration, and investigate its potential mechanisms. Exposure to different concentrations of PTC (0-500 µM) significantly inhibited the cell viability and migration ability (5 µM PTC exposure), and also caused the cell cycle arrest at the lowest dose (1 µM PTC exposure). Transcriptome analysis revealed that PTC exposure disturbed multiple biological processes including cell cycle and apoptosis, consistent with cell phenotype. Specifically, eukaryotic translation initiation factor 4E binding protein 1 (EIF4EBP1, 4E-BP1) was identified as up-regulated in PTC exposure group and knockdown of EIF4EBP1, and attenuated the G1 phase arrest induced by PTC exposure. In summary, our data demonstrated that 4E-BP1 participated in PTC-induced cell cycle arrest in extravillous trophoblast cells by regulating cyclin D1. These findings shed light on the potential adverse effect of PTC exposure on the embryo implantation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/genética , Triazóis/toxicidade , Trofoblastos/efeitos dos fármacos , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ciclina D1/metabolismo , Relação Dose-Resposta a Droga , Feminino , Fungicidas Industriais/administração & dosagem , Fungicidas Industriais/toxicidade , Técnicas de Silenciamento de Genes , Humanos , Triazóis/administração & dosagem , Trofoblastos/citologia , Regulação para Cima/efeitos dos fármacos
10.
Bioengineered ; 13(1): 206-216, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34964705

RESUMO

Multiple studies have confirmed that adipokines are compactly relevant to insulin resistance and participate in the pathogenesis of gestational diabetes mellitus (GDM). This paper aimed to study the effects of C1q/tumor necrosis factor related protein (CTRP)6 on the phenotypes of trophoblast cells, covering cell proliferation, invasion and migration, and initially explore the mechanism. High glucose was used to induce trophoblast cells to establish an in vitro model. The expression levels of CTRP6 were firstly determined, and then the effects of CTRP6 knockdown on cell viability, apoptosis, migration and invasion were assessed using CCK8, TUNEL, wound healing, Transwell assays. Moreover, the role of peroxisome proliferator-activated receptor gamma (PPARγ), probable target of CTRP6, was evaluated through co-transfection with PPARγ overexpression vector. The results of the present study revealed that CTRP6 and PPARγ were both upregulated in high glucose-induced cells. And CTRP6 knockdown could significantly elevate the abilities of cell viability, migration and invasion, and avoid cell apoptosis. In addition, PPARγ overexpression was found to restrain the protective effects of CTRP6 knockdown on the above aspects, indicating CTRP6 played a role in trophoblast cells via inhibiting PPARγ expression. In conclusion, CTRP6 regulated the viability, migration and invasion of high glucose-induced gestational trophoblast cells through PPARγ signaling.


Assuntos
Colágeno/metabolismo , Glucose/efeitos adversos , PPAR gama/metabolismo , Trofoblastos/citologia , Regulação para Cima , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Colágeno/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , PPAR gama/genética , Fenótipo , Transdução de Sinais/efeitos dos fármacos , Trofoblastos/efeitos dos fármacos , Trofoblastos/metabolismo
11.
Bioengineered ; 13(1): 319-330, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34969354

RESUMO

Gestational diabetes mellitus (GDM) is a prevalent and risky pregnant complication which warrants targeted therapy for restriction the inflammation and apoptosis of trophoblast cells. This study sought to analyze the aberrant expression and regulatory mechanism of microRNA (miR)-134-5p in GDM. The miR-134-5p expression in the serum of GDM patients and normal participants was detected via qRT-PCR, followed by receiver operating characteristic (ROC) curve analysis. In vitro GDM cell model was established in the HTR-8/SVneo cells using 25 mmol/L glucose, followed by transfection with miR-134-5p inhibitor and si-Forkhead box p2(FOXP2). The miR-134-5p and FOXP2 expressions, TNF-α, IL-1ß, and IL-10 levels, cell proliferation, migration, and apoptosis were determined by a combination of qRT-PCR, western blot, ELISA, and cell counting Kit-8, Transwell assay, and flow cytometry. The binding relationship between miR-134-5p and FOXP2 was predicted and verified. Our results revealed that miR-134-5p was increased in the serum of GDM patients and could serve as a critical diagnostic marker for GDM. Moreover, miR-134-5p was upregulated in the high glucose (HG)-induced HTR-8/SVneo cells. The miR-134-5p inhibition suppressed the inflammation and apoptosis of HG-induced HTR-8/SVneo cells. miR-134-5p inhibited FOXP2 expression. FOXP2 expression was decreased in GDM. FOXP2 inhibition attenuated the function of miR-134-5p in HG-induced HTR-8/SVneo cells. Overall, miR-134-5p inhibited the FOXP2 expression to facilitate the inflammation and apoptosis of trophoblast cells, thereby exacerbating GDM.


Assuntos
Diabetes Gestacional/diagnóstico , Fatores de Transcrição Forkhead/genética , Glucose/efeitos adversos , MicroRNAs/sangue , Trofoblastos/citologia , Regulação para Cima , Adulto , Estudos de Casos e Controles , Linhagem Celular , Movimento Celular , Proliferação de Células , Diabetes Gestacional/sangue , Diabetes Gestacional/genética , Feminino , Marcadores Genéticos , Humanos , Idade Materna , Modelos Biológicos , Gravidez , Curva ROC , Trofoblastos/efeitos dos fármacos , Trofoblastos/metabolismo
12.
Toxicology ; 465: 153060, 2022 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-34871708

RESUMO

With the increasing application of cell culture models as primary tools for predicting chemical safety, the quantitative extrapolation of the effective dose from in vitro to in vivo (QIVIVE) is of increasing importance. For developmental toxicity this requires scaling the in vitro observed dose-response characteristics to in vivo fetal exposure, while integrating maternal in vivo kinetics during pregnancy, in particular transplacental transfer. Here the transfer of substances across the placental barrier, has been studied using the in vitro BeWo cell assay and six embryotoxic compounds of different kinetic complexity. The BeWo assay results were incorporated in an existing generic Physiologically Based Kinetic (PBK) model which for this purpose was extended with rat pregnancy. Finally, as a "proof of principle", the BeWo PBK model was used to perform a QIVIVE based on developmental toxicity as observed in various different in vitro toxicity assays. The BeWo results illustrated different transport profiles of the chemicals across the BeWo monolayer, allocating the substances into two distinct groups: the 'quickly-transported' and the 'slowly-transported'. BeWo PBK exposure simulations during gestation were compared to experimentally measured maternal blood and fetal concentrations and a reverse dosimetry approach was applied to translate in vitro observed embryotoxicity into equivalent in vivo dose-response curves. This approach allowed for a direct comparison of the in vitro dose-response characteristics as observed in the Whole Embryo Culture (WEC), and the Embryonic Stem Cell test (cardiac:ESTc and neural:ESTn) with in vivo rat developmental toxicity data. Overall, the in vitro to in vivo comparisons suggest a promising future for the application of such QIVIVE methodologies for screening and prioritization purposes of developmental toxicants. Nevertheless, the clear need for further improvements is acknowledged for a wider application of the approach in chemical safety assessment.


Assuntos
Troca Materno-Fetal , Modelos Biológicos , Testes de Toxicidade , Trofoblastos/efeitos dos fármacos , Animais , Transporte Biológico , Biomarcadores/sangue , Caproatos/toxicidade , Linhagem Celular , Relação Dose-Resposta a Droga , Feminino , Sangue Fetal/metabolismo , Idade Gestacional , Glicolatos/toxicidade , Humanos , Miconazol/toxicidade , Permeabilidade , Ácidos Ftálicos/toxicidade , Gravidez , Estudo de Prova de Conceito , Ratos , Reprodutibilidade dos Testes , Medição de Risco , Silanos/toxicidade , Toxicocinética , Triazóis/toxicidade , Trofoblastos/metabolismo , Trofoblastos/patologia , Ácido Valproico/toxicidade
13.
Placenta ; 117: 95-108, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34785431

RESUMO

INTRODUCTION: Abnormal extravillous trophoblast (EVT) function is closely related to preeclampsia (PE) and may be caused by inadequate autophagy, apoptosis, and senescence. Cyclosporin A (CsA) is an effective immunosuppressant that has been reported to stimulate autophagy and exert benign biological effects on EVTs. Therefore, we hypothesized that CsA may display therapeutic efficacy against PE by activating autophagy. METHODS: We established the nitro-l-arginine methyl ester (l-NAME)-induced preeclamptic mice model and a hypoxia-reoxygenation (H/R) model in vitro. The effects of CsA on autophagy were evaluated by western blotting (WB). The effects of CsA on apoptosis were analyzed by Hematoxylin-eosin (H&E) staining, cell apoptosis assay and WB. Senescence-associated ß-galactosidase (SA-ß-gal) staining, RT-qPCR and WB were used to examine the senescence level. RT-qPCR were used to detect the senescence-associated secretory phenotype (SASP) level. DCFH-DA fluorescent probe, dihydroethidium (DHE) staining and mitochondrial membrane potential (ΔΨm) were used to detect senescence-associated mitochondrial dysfunction (SAMD). RESULTS: CsA alleviated PE-like symptoms and reduced placental necrosis and senescence in mice injected with l-NAME. CsA ameliorated placental SASP and SAMD level induced by l-NAME. CsA also upregulated the expression of autophagic proteins in mouse placentas disrupted using l-NAME. In vitro, we found that CsA reversed H/R-induced apoptosis and senescence, as well as decreasing SASP and SAMD levels and upregulating autophagic proteins levels. Notably, 3-methyladenine (3-MA), an early phase inhibitor of autophagosome formation, abolished the protective effects of CsA against H/R. DISCUSSION: CsA may display some therapeutic effects against PE by activating autophagy in vivo and in vitro.


Assuntos
Autofagia/efeitos dos fármacos , Ciclosporina/uso terapêutico , Imunossupressores/uso terapêutico , Pré-Eclâmpsia/tratamento farmacológico , Trofoblastos/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Ciclosporina/farmacologia , Avaliação Pré-Clínica de Medicamentos , Feminino , Imunossupressores/farmacologia , Camundongos , NG-Nitroarginina Metil Éster , Placenta/patologia , Pré-Eclâmpsia/patologia , Gravidez , Fenótipo Secretor Associado à Senescência
14.
Bioengineered ; 13(1): 395-406, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34787071

RESUMO

Gestational diabetes mellitus (GDM) is a situation where glucose intolerance is found in pregnant women without a previous diagnosis of diabetes. The role of Kruppel-like factor 9 (KLF9) has not been investigated in GDM, which constituted the aim of our study. HTR8/SVneo cells were induced by high glucose (HG) and pregnant mice were treated with streptozocin (STZ) to establish GDM model in vitro and in vivo, respectively. The expression level of KLF9 was detected by real-time PCR, immunohistochemical staining, and Western blot. Cell viability, apoptosis, inflammation, and oxidative stress were investigated by cell counting kit-8 (CCK-8), TUNEL, enzyme-linked immunosorbent assay (ELISA) and oxidative stress detection kits, respectively. The interaction of KLF9 with dimethylarginine dimethylaminohydrolase 2 (DDAH2) was predicted by bioinformatic tools and confirmed by luciferase reporter assay and chromatin immunoprecipitation (ChIP). The expression of KLF9 was increased in the placental tissues of GDM patients and HG-induced HTR8/SVneo cells. Silencing of KLF9 increased cell viability, reduced cell apoptosis, and suppressed inflammation and oxidative stress in HG-induced HTR8/SVneo cells. KLF9 could bind to DDAH2 promoter and negatively regulate DDAH2 expression. Inhibition of DDAH2 partly weakened the effects of KLF9 silencing on cell apoptosis, inflammation, and oxidative stress. The suppressive effects of KLF9 silencing on blood glucose and insulin concentration in vivo were also abolished by DDAH2 knockdown. In conclusion, we provided evidence that interference of KLF9 could hinder the development of GDM by alleviating cell apoptosis, inflammation, and oxidative stress through upregulating DDAH2, which might instruct the targeting therapies against GDM.Abbreviations: KLF9: Kruppel-like factor 9; DDAH2: dimethylarginine dimethylaminohydrolase 2 ; GDM: gestational diabetes mellitus; ELISA: enzyme-linked immunosorbent assay; CCK-8: cell counting kit-8; ChIP: chromatin immunoprecipitation; sh: short hairpin; HG: high glucose; PBS: phosphate-buffered saline; DAPI: 4, 6-diamidino-2-phenylindole; IL-6: Interleukin-6; TNF-α: tumor necrosis factor-α; ROS: reactive oxygen species; MDA: malondialdehyde; SOD: superoxide dismutase; wt: wild-type; mut: mutant.


Assuntos
Amidoidrolases/metabolismo , Diabetes Gestacional/genética , Glucose/efeitos adversos , Fatores de Transcrição Kruppel-Like/genética , Estreptozocina/efeitos adversos , Regulação para Cima , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Diabetes Gestacional/induzido quimicamente , Diabetes Gestacional/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Gravidez , Trofoblastos/citologia , Trofoblastos/efeitos dos fármacos , Trofoblastos/metabolismo
15.
Reprod Toxicol ; 107: 60-68, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34838982

RESUMO

During pregnancy, the migration and invasion of extravillous trophoblasts (EVTs) into the maternal uterus is essential for proper development of the placenta and fetus. During the first trimester, EVTs engraft and remodel maternal spiral arteries allowing for efficient blood flow and the transfer of essential nutrients and oxygen to the fetus. Aberrant migration of EVTs leading to either shallow or deep invasion into the uterus has been implicated in a number of gestational pathologies including preeclampsia, fetal growth restriction, and placenta accreta spectrum. The migration and invasion of EVTs is well-coordinated to ensure proper placentation. However, recent data point to the ability of xenobiotics to disrupt EVT migration. These xenobiotics include heavy metals, endocrine disrupting chemicals, and organic contaminants and have often been associated with adverse pregnancy outcomes. In most instances, xenobiotics appear to reduce EVT migration; however, there are select examples of enhanced motility after chemical exposure. In this review, we provide an overview of the 1) current experimental approaches used to evaluate EVT migration and invasion in vitro, 2) ability of environmental chemicals and pharmaceuticals to enhance or retard EVT motility, and 3) signaling pathways responsible for altered EVT migration that are sensitive to disruption by xenobiotics.


Assuntos
Movimento Celular/efeitos dos fármacos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Poluentes Ambientais/toxicidade , Trofoblastos/efeitos dos fármacos , Xenobióticos/toxicidade , Animais , Humanos , Trofoblastos/fisiologia
16.
Reprod Sci ; 29(1): 163-172, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34382203

RESUMO

Preeclampsia (PE) is a life-threatening pregnancy complication associated with diminished trophoblast migration and invasion. Wnt signalling is one of the most important regulators of placentation. Secreted frizzled-related protein 5 (SFRP5) is an anti-inflammatory adipokine that may inhibit Wnt signalling. In this study, we aimed to investigate the relationship between SFRP5 and PE and its effect on trophoblast function, as well as the underlying signalling pathways. SFRP5 levels in the serum and placental tissues were detected using enzyme-linked immunosorbent assay and immunohistochemistry, respectively. To evaluate the effect of SFRP5 on Wnt signalling, the human trophoblast cell line HTR8/SVneo was treated with recombinant human SFRP5 and Dickkopf-related protein 1 (Dkk-1, canonical Wnt inhibitor) proteins and lithium chloride (LiCl, canonical Wnt agonist). The migration and invasion ability of HTR8/SVneo cells was evaluated using wound-healing and Matrigel Transwell assays. The activities of multiple matrix metalloproteinases (MMP)-2/9 were detected using gelatin zymography. Expression of glycogen synthase kinase-3 beta (GSK3ß) and ß-catenin proteins was investigated using western blotting. The serum SFRP5 levels were elevated in patients with PE, but SFRP5 expression was not detected in the placental tissues. Furthermore, SFRP5 inhibited the migration and invasion of HTR8/SVneo cells in vitro, increased GSK3ß, and decreased ß-catenin expression and MMP-2/9 activity in HTR8/SVneo cells. In conclusion, this study suggests that SFRP5 inhibits trophoblast migration and invasion potentially via the inhibition of Wnt/ß-catenin signalling, which might be involved in the development of PE. However, the primary cause of the increased SFRP5 levels needs to be investigated.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/sangue , Pré-Eclâmpsia/sangue , Trofoblastos/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal/farmacologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Placenta/efeitos dos fármacos , Placenta/metabolismo , Gravidez , Trofoblastos/efeitos dos fármacos , beta Catenina/metabolismo
17.
Biochim Biophys Acta Mol Cell Res ; 1869(1): 119139, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34624436

RESUMO

Trophoblasts are specialized epithelial cells of the placenta that are involved in invasion, communication and the exchange of materials between the mother and fetus. Cytoplasmic Ca2+ ([Ca2+]c) plays critical roles in regulating such processes in other cell types, but relatively little is known about the mechanisms that control this second messenger in trophoblasts. In the current study, the presence of RyRs and their accessory proteins in placental tissues and in the BeWo choriocarcinoma, a model trophoblast cell-line, were examined using immunohistochemistry and Western immunoblotting. Contributions of RyRs to Ca2+ signalling and to random migration in BeWo cells were investigated using fura-2 fluorescent and brightfield videomicroscopy. The effect of RyR inhibition on reorganization of the F-actin cytoskeleton elicited by the hormone angiotensin II, was determined using phalloidin-labelling and confocal microscopy. RyR1 and RyR3 proteins were detected in trophoblasts of human first trimester and term placental villi, along with the accessory proteins triadin and calsequestrin. Similarly, RyR1, RyR3, triadin and calsequestrin were detected in BeWo cells. In this cell-line, activation of RyRs with micromolar ryanodine increased [Ca2+]c, whereas pharmacological inhibition of these channels reduced Ca2+ transients elicited by the peptide hormones angiotensin II, arginine vasopressin and endothelin 1. Angiotensin II increased the velocity, total distance and Euclidean distance of random migration by BeWo cells and these effects were significantly reduced by tetracaine and by inhibitory concentrations of ryanodine. RyRs contribute to reorganization of the F-actin cytoskeleton elicited by angiotensin II, since inhibition of these channels restores the parallelness of these structures to control levels. These findings demonstrate that trophoblasts contain a suite of proteins similar to those in other cell types possessing highly developed Ca2+ signal transduction systems, such as skeletal muscle. They also indicate that these channels regulate the migration of trophoblast cells, a process that plays a key role in development of the placenta.


Assuntos
Sinalização do Cálcio , Movimento Celular , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Trofoblastos/metabolismo , Citoesqueleto de Actina/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Hormônios Peptídicos/farmacologia , Trofoblastos/efeitos dos fármacos , Trofoblastos/fisiologia
18.
J Nutr Biochem ; 99: 108866, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34563666

RESUMO

Palmitic acid (PA) induces apoptosis in the human trophoblast cell line HTR8/SVneo. However, the molecular mechanism underlying this effect remains unclear. Although small noncoding RNAs are involved in trophoblast growth and invasion during early pregnancy, the functional roles of tRNA-derived species are currently unknown. Therefore, the purpose of this study was to examine the involvement of tRNA-derived species in PA-induced apoptosis in human trophoblasts. In this study, we investigate the expression and function of tRNA-derived stress-induced RNAs (tiRNAs) in HTR8/SVneo. We determined the expression of tiRNAs in HTR8/SVneo cells in response to PA. Then, we transfected inhibitor of target tiRNA in HTR8/SVneo with or without PA to examine the tRNA-derived species-regulated intracellular signal transduction by detecting calcium homeostasis, mitochondrial membrane potential, and signaling proteins. We found that the expression of tRNAGly-derived tiRNAs decreased in PA-treated human trophoblasts. Moreover, inhibition of tiRNAGlyCCC/GCC enhanced the PA-induced apoptosis along with the induction of DNA fragmentation and mitochondrial depolarization. Inhibition of tiRNAGlyCCC/GCC enhanced the expression of endoplasmic reticulum stress-related proteins and increased Ca2+ levels in the cytoplasm and mitochondria. Moreover, the levels of cytochrome c released from the mitochondria were synergistically affected by tiRNAGlyCCC/GCC inhibitor and PA. Furthermore, artificial regulation of ANG inhibited the expression of tiRNAGlyCCC/GCC and similar effects were observed upon the inhibition of tiRNAGlyCCC/GCC in human trophoblasts. These results suggest that tiRNAGlyCCC/GCC might be the molecule via which PA induces its effects in human trophoblasts.


Assuntos
Apoptose/efeitos dos fármacos , Ácido Palmítico/efeitos adversos , RNA de Transferência de Glicina/metabolismo , Trofoblastos/citologia , Cálcio/metabolismo , Fragmentação do DNA/efeitos dos fármacos , Humanos , RNA de Transferência de Glicina/genética , Trofoblastos/efeitos dos fármacos , Trofoblastos/metabolismo
19.
Bioengineered ; 12(2): 12789-12799, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34895049

RESUMO

Gestational diabetes mellitus (GDM) is a common disease in pregnant women, imposing risks on both mother and fetus. Dysregulated nesfatin-1 has been observed in women with GDM, but the specific role of nesfatin-1 underlying the pathological process of GDM is unclear. The main objective of this study is to investigate the role and the molecular mechanism of nesfatin-1 in GDM. HTR-8/SVneo cells were treated with high glucose (HG)/high lipid (HL) to mimic the injured trophoblast of GDM in vitro. Cell viability, cytotoxicity and apoptosis were measured using CCK-8, LDH and TUNEL assays, respectively. The levels of inflammatory cytokines and antioxidant factors were detected using their commercial kits. ATP level and cytochrome c were determined with corresponding detecting kits. Quantitative real-time PCR and Western blot were performed to detect the expression of corresponding genes. The results showed that nesfatin-1 was downregulated upon HG/HL stimulation. Nesfatin-1 treatment greatly alleviated HG/HL-induced cell viability loss, cytotoxicity, inflammatory response, oxidative stress, and apoptosis in HTR-8/SVneo cells. In addition, nesfatin-1 promoted ATP generation, reduced the leakage of cytochrome c from mitochondria to cytoplasm, and upregulated mitochondrial transcription factor A (TFAM) and nuclear respiratory factor 1 (NRF1), alleviating mitochondrial dysfunction. Furthermore, nesfatin-1 inhibited p38 MAPK signaling. p79350, an agonist of p38 MAPK signaling, remarkably hindered the protective role of nesfatin-1 in HG/HL-induced HTR-8/SVneo cells. In conclusion, nesfatin-1 exerted a protective effect on GDM model in vitro, by regulating p38 MAPK signaling pathway, providing novel insights of treating GDM.


Assuntos
Diabetes Gestacional/patologia , Glucose/toxicidade , Lipídeos/toxicidade , Nucleobindinas/farmacologia , Trofoblastos/patologia , Apoptose/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Feminino , Humanos , Inflamação/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Gravidez , Substâncias Protetoras/farmacologia , Trofoblastos/efeitos dos fármacos , Trofoblastos/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
20.
Environ Health Perspect ; 129(11): 117001, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34747641

RESUMO

BACKGROUND: Our environment is replete with chemicals that can affect embryonic and extraembryonic development. Dioxins, such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), are compounds affecting development through the aryl hydrocarbon receptor (AHR). OBJECTIVES: The purpose of this investigation was to examine the effects of TCDD exposure on pregnancy and placentation and to evaluate roles for AHR and cytochrome P450 1A1 (CYP1A1) in TCDD action. METHODS: Actions of TCDD were examined in wild-type and genome-edited rat models. Placenta phenotyping was assessed using morphological, biochemical, and molecular analyses. RESULTS: TCDD exposures were shown to result in placental adaptations and at higher doses, pregnancy termination. Deep intrauterine endovascular trophoblast cell invasion was a prominent placentation site adaptation to TCDD. TCDD-mediated placental adaptations were dependent upon maternal AHR signaling but not upon placental or fetal AHR signaling nor the presence of a prominent AHR target, CYP1A1. At the placentation site, TCDD activated AHR signaling within endothelial cells but not trophoblast cells. Immune and trophoblast cell behaviors at the uterine-placental interface were guided by the actions of TCDD on endothelial cells. DISCUSSION: We identified an AHR regulatory pathway in rats activated by dioxin affecting uterine and trophoblast cell dynamics and the formation of the hemochorial placenta. https://doi.org/10.1289/EHP9256.


Assuntos
Dioxinas , Placentação , Dibenzodioxinas Policloradas , Receptores de Hidrocarboneto Arílico , Animais , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/metabolismo , Dioxinas/toxicidade , Células Endoteliais/metabolismo , Feminino , Placenta/metabolismo , Dibenzodioxinas Policloradas/toxicidade , Gravidez , Ratos , Receptores de Hidrocarboneto Arílico/metabolismo , Trofoblastos/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...