Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 407
Filtrar
1.
Tissue Cell ; 74: 101698, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34871824

RESUMO

To establish a functional placenta, its development needs adequate trophoblastic invasiveness. The intricate and complex morphological and molecular aspects regulating trophoblastic invasion during endotheliochorial placentation of domestic carnivores and their similarities and differences with the hemochorial placenta are still poorly understood. During placentation processes, from the time of implantation, trophoblast cells invade the uterine endometrium where they achieve extensive degradation and remodeling of extracellular matrix components; in this process, matrix metalloproteinases (MMPs), particularly MMP-2 and 9, have an essential role in rebuilding, cell migration, and invasiveness. This review provides an overview of comparative trophoblast invasive events and the expression and activity of MMP-2 and 9 during endotheliochorial and hemochorial placentation, emphasizing dog and mouse placental models. Understanding of trophoblastic invasiveness in two models of placentation, the intermediately invasive domestic carnivore endotheliochorial placenta, and the more highly invasive mouse hemochorial placenta, contributes to deepen knowledge of the trophoblast invasive processes and their diverse and complex human placental alterations, such as preeclampsia.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Metaloproteinase 2 da Matriz/biossíntese , Metaloproteinase 9 da Matriz/biossíntese , Placentação , Trofoblastos/enzimologia , Animais , Cães , Endométrio/enzimologia , Feminino , Humanos , Camundongos , Gravidez
2.
Placenta ; 117: 187-193, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34929459

RESUMO

INTRODUCTION: Recent evidence supports the - rare - occurrence of vertical transplacental SARS-CoV-2 transmission. We previously determined that placental expression of angiotensin-converting enzyme 2 (ACE2), the SARS-CoV-2 receptor, and associated viral cell entry regulators is upregulated by hypoxia. In the present study, we utilized a clinically relevant model of SARS-CoV-2-associated chronic histiocytic intervillositis/massive perivillous fibrin deposition (CHIV/MPFVD) to test the hypothesis that placental hypoxia may facilitate placental SARS-CoV-2 infection. METHODS: We performed a comparative immunohistochemical and/or RNAscope in-situ hybridization analysis of carbonic anhydrase IX (CAIX, hypoxia marker), ACE2 and SARS-CoV-2 expression in free-floating versus fibrin-encased chorionic villi in a 20-weeks' gestation placenta with SARS-CoV-2-associated CHIV/MPVFD. RESULTS: The levels of CAIX and ACE2 immunoreactivity were significantly higher in trophoblastic cells of fibrin-encased villi than in those of free-floating villi, consistent with hypoxia-induced ACE2 upregulation. SARS-CoV-2 showed a similar preferential localization to trophoblastic cells of fibrin-encased villi. DISCUSSION: The localization of SARS-CoV-2 to hypoxic, fibrin-encased villi in this placenta with CHIV/MPVFD suggests placental infection and, therefore, transplacental SARS-CoV-2 transmission may be promoted by hypoxic conditions, mediated by ACE2 and similar hypoxia-sensitive viral cell entry mechanisms. Understanding of a causative link between placental hypoxia and SARS-CoV-2 transmittability may potentially lead to the development of alternative strategies for prevention of intrauterine COVID-19 transmission.


Assuntos
COVID-19/complicações , Fibrina/análise , Hipóxia/virologia , Placenta/virologia , Complicações Infecciosas na Gravidez/virologia , SARS-CoV-2/isolamento & purificação , Adulto , Enzima de Conversão de Angiotensina 2/análise , COVID-19/patologia , COVID-19/virologia , Anidrase Carbônica IX/análise , Vilosidades Coriônicas/enzimologia , Vilosidades Coriônicas/virologia , Feminino , Idade Gestacional , Histiócitos/patologia , Humanos , Hipóxia/patologia , Transmissão Vertical de Doenças Infecciosas , Necrose/virologia , Placenta/química , Placenta/patologia , Gravidez , Natimorto , Trofoblastos/enzimologia , Trofoblastos/virologia
3.
Sci Rep ; 11(1): 16595, 2021 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-34400721

RESUMO

Fetal growth restriction is a leading cause of stillbirth that often remains undetected during pregnancy. Identifying novel biomarkers may improve detection of pregnancies at risk. This study aimed to assess syndecan-1 as a biomarker for small for gestational age (SGA) or fetal growth restricted (FGR) pregnancies and determine its molecular regulation. Circulating maternal syndecan-1 was measured in several cohorts; a large prospective cohort collected around 36 weeks' gestation (n = 1206), a case control study from the Manchester Antenatal Vascular service (285 women sampled at 24-34 weeks' gestation); two prospective cohorts collected on the day of delivery (36 + 3-41 + 3 weeks' gestation, n = 562 and n = 405 respectively) and a cohort who delivered for preterm FGR (< 34 weeks). Circulating syndecan-1 was consistently reduced in women destined to deliver growth restricted infants and those delivering for preterm disease. Syndecan-1 secretion was reduced by hypoxia, and its loss impaired proliferation. Matrix metalloproteinases and mitochondrial electron transport chain inhibitors significantly reduced syndecan-1 secretion, an effect that was rescued by coadministration of succinate, a mitochondrial electron transport chain activator. In conclusion, circulating syndecan-1 is reduced among cases of term and preterm growth restriction and has potential for inclusion in multi-marker algorithms to improve detection of poorly grown fetuses.


Assuntos
Retardo do Crescimento Fetal/sangue , Metaloproteinases da Matriz/fisiologia , Mitocôndrias/fisiologia , Placenta/metabolismo , Complicações na Gravidez/sangue , Sindecana-1/sangue , Adulto , Área Sob a Curva , Peso ao Nascer , Hipóxia Celular , Parto Obstétrico , Diabetes Gestacional/sangue , Transporte de Elétrons/efeitos dos fármacos , Feminino , Idade Gestacional , Humanos , Hipertensão/sangue , Recém-Nascido , Recém-Nascido Pequeno para a Idade Gestacional , Metformina/farmacologia , Mitocôndrias/efeitos dos fármacos , Tamanho do Órgão , Sobrepeso/sangue , Pré-Eclâmpsia/sangue , Gravidez , Curva ROC , Fumar/sangue , Trofoblastos/enzimologia
4.
Cell Death Dis ; 12(7): 635, 2021 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-34155192

RESUMO

FURIN is a pro-protein convertase previously shown to be important for placental syncytialisation (Zhou et al. [1]), a process of cell fusion whereby placental cytotrophoblast cells fuse to form a multinucleated syncytium. This finding has been broadly accepted however, we have evidence suggesting the contrary. Spontaneously syncytialising term primary human trophoblast cells and BeWo choriocarcinoma cells were treated with either FURIN siRNA or negative control siRNA or the protease inhibitor, DEC-RVKR-CMK, or vehicle. Cells were then left to either spontaneously syncytialise (primary trophoblasts) or were induced to syncytialise with forskolin (BeWo). Effects on syncytialisation were measured by determining human chorionic gonadotrophin secretion and E-cadherin protein levels. We showed that FURIN is not important for syncytialisation in either cell type. However, in primary trophoblasts another protease also inhibited by DEC-RVKR-CMK, may be involved. Our results directly contrast with those published by Zhou et al. Zhou et al. however, used first trimester villous explants to study syncytialisation, and we used term primary trophoblasts. Therefore, we suggest that FURIN may be involved in syncytialisation of first trimester trophoblasts, but not term trophoblasts. What is more concerning is that our results using BeWo cells do not agree with their results, even though for the most part, we used the same experimental design. It is unclear why these experiments yielded different results, however we wanted to draw attention to simple differences in measuring syncytialisation or flaws in method reporting (including omission of cell line source and passage numbers, siRNA concentration and protein molecular weights) and choice of immunoblot loading controls, that could impact on experimental outcomes. Our study shows that careful reporting of methods by authors and thorough scrutiny by referees are vital. Furthermore, a universal benchmark for measuring syncytialisation is required so that various studies of syncytialisation can be validated.


Assuntos
Fusão Celular , Furina/metabolismo , Placentação , Trofoblastos/enzimologia , Clorometilcetonas de Aminoácidos/farmacologia , Antígenos CD/metabolismo , Caderinas/metabolismo , Linhagem Celular Tumoral , Gonadotropina Coriônica/metabolismo , Colforsina/farmacologia , Feminino , Furina/antagonistas & inibidores , Furina/genética , Humanos , Placentação/efeitos dos fármacos , Gravidez , Primeiro Trimestre da Gravidez , Inibidores de Serina Proteinase/farmacologia , Nascimento a Termo , Trofoblastos/efeitos dos fármacos
5.
Am J Physiol Regul Integr Comp Physiol ; 320(5): R653-R662, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33621475

RESUMO

Currently, there is no effective treatment for placental dysfunction in utero. In a ligated mouse model of fetal growth restriction (FGR), nanoparticle-mediated human insulin-like 1 growth factor (hIGF1) gene delivery (NP-Plac1-hIGF1) increased hIGF1 expression and maintained fetal growth. However, whether it can restore fetal growth remains to be determined. Using the endothelial nitric oxide synthase knockout (eNOS-/-) mouse model, a genetic model of FGR, we found that despite inducing expression of hIGF1 in the placentas treated with NP-Plac1-hIGF1 (P = 0.0425), FGR did not resolve. This was associated with no change to the number of fetal capillaries in the placental labyrinth; an outcome which was increased with NP-Plac1-hIGF1 treatment in the ligated mouse model, despite increased expression of angiopoietin 1 (P = 0.05), and suggested IGF1 signaling in the placenta requires eNOS to modulate placenta angiogenesis. To further assess this hypothesis, BeWo choriocarcinoma cell line and human placental explant cultures were treated with NP-Plac1-hIGF1, oxidative stress was induced with hydrogen peroxide (H2O2), and NOS activity was inhibited using the inhibitor NG-monomethyl-l-arginine (l-NMMA). In both BeWo cells and explants, the protective effect of NP-Plac1-hIGF1 treatment against H2O2-induced cell death/lactate dehydrogenase release was prevented by eNOS inhibition (P = 0.003 and P < 0.0001, respectively). This was associated with an increase in mRNA expression of oxidative stress markers hypoxia inducing factor 1α (HIF1α; P < 0.0001) and ADAM10 (P = 0.0002) in the NP-Plac1-hIGF1 + H2O2 + l-NMMA-treated BeWo cells. These findings show for the first time the requirement of eNOS/NOS in IGF1 signaling in placenta cells that may have implications for placental angiogenesis and fetal growth.


Assuntos
Retardo do Crescimento Fetal/terapia , Feto/irrigação sanguínea , Terapia Genética , Fator de Crescimento Insulin-Like I/metabolismo , Neovascularização Fisiológica , Óxido Nítrico Sintase Tipo III/metabolismo , Placenta/irrigação sanguínea , Trofoblastos/enzimologia , Proteína ADAM10/genética , Proteína ADAM10/metabolismo , Secretases da Proteína Precursora do Amiloide/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Desenvolvimento Fetal , Retardo do Crescimento Fetal/enzimologia , Retardo do Crescimento Fetal/genética , Retardo do Crescimento Fetal/fisiopatologia , Técnicas de Transferência de Genes , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fator de Crescimento Insulin-Like I/genética , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nanopartículas , Óxido Nítrico Sintase Tipo III/genética , Estresse Oxidativo , Gravidez , Transdução de Sinais , Técnicas de Cultura de Tecidos , Trofoblastos/patologia
6.
Mol Hum Reprod ; 27(3)2021 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-33629098

RESUMO

Extravillous trophoblast cell (EVT) invasion is tightly controlled, and its dysregulation can lead to altered spiral artery remodeling and contribute to a number of different pregnancy complications. Angiopoietin-2 (Ang-2) is expressed by trophoblast cells and various cells in the decidua, and trophoblast cells express its receptor, Tie2. Ang-2 has been shown to play roles in tumor progression and metastasis but it is not known if it also regulates EVT invasion. Here, we show that both the HTR-8/SVneo cell line and primary isolates of human EVT expressed various integrins and the Tie2 receptor, and Ang-2 stimulated their migration and/or invasion. Ang-2 increased expression of matrix metalloproteinase (MMP)2 and MMP9, altered the cytoskeleton of HTR-8/SVneo cells and also induced phosphorylation of Tie2, JNK and c-Jun. Inhibition of p-JNK (using SP600125) blocked the Ang-2 induced invasion of HTR-8/SVneo cells. In addition, inhibition of Tie2 (pexmetinib) and integrin signaling (RGDS and ATN-161) also blocked Ang-2-induced invasion. In conclusion, we demonstrate that Ang-2 can stimulate EVT invasion via a mechanism associated with activation of both the Tie2 receptor and integrins, which appear to work through different pathways; Tie2 through the JNK/c-JUN pathway and integrins through an as yet unidentified pathway(s). We therefore propose that any alterations in Ang-2 expression in the decidua would lead to an imbalance in pro- and anti-invasive factors, disrupting regulation of EVT invasion and spiral artery remodeling and thereby contribute to the etiology of several complications of pregnancy.


Assuntos
Angiopoietina-2/farmacologia , Movimento Celular/efeitos dos fármacos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Transdução de Sinais/efeitos dos fármacos , Trofoblastos/efeitos dos fármacos , Linhagem Celular , Feminino , Humanos , Integrinas/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Fosforilação , Gravidez , Complicações na Gravidez/enzimologia , Proteínas Proto-Oncogênicas c-jun/metabolismo , Receptor TIE-2/agonistas , Receptor TIE-2/metabolismo , Trofoblastos/enzimologia
7.
Reprod Sci ; 28(2): 550-561, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32974855

RESUMO

Preeclampsia (PE) is a leading cause of perinatal and maternal mortality. Considering that Nesfatin-1 was reported to be downregulated in serum of PE patients, we aimed to explore the functional role of Nesfatin-1 in trophoblast cells. Cell transfection was conducted to overexpress or inhibit Nesfatin-1, and its expression was measured by quantitative PCR. Cell proliferation, migration, and invasion abilities were determined employing CCK-8, flow cytometry, wound-healing, and transwell assays. Immunofluorescence assay was performed to detect E-cadherin and vimentin. ROS, MDA, and SOD levels were measured using their corresponding commercial kits. Western blot was used to identify the expression of vital kinases in PI3K/AKT/mTOR or GSK3ß pathway and invasion-related proteins in trophoblast cells. Nesfatin-1 knockdown significantly suppressed proliferation, migration, and invasion and increased cell arrest in G1 phase, as well as downregulated expressions of MMP2/9 in HTR-8/SVneo cells. Besides, Nesfatin-1 knockdown promoted the expression of E-cadherin and reduced the expression of vimentin. Additionally, the levels of ROS, MDA, and SOD were elevated upon Nesfatin-1 knockdown. On the contrary, Nesfatin-1 overexpression exerted the opposite effects. Nesfatin-1 promoted the activation of PI3K/AKT/mTOR or GSK3ß pathway, blocking of which reversed the promotive effects on trophoblast invasion and the inhibitory effects on oxidative stress of Nesfatin-1 in HTR-8/SVneo cells. In short, this study revealed that Nesfatin-1 promoted trophoblast cell proliferation, migration, invasion, and EMT and suppressed oxidative stress by activating PI3K/AKT/mTOR and AKT/GSK3ß signaling pathway, laying the foundation for the development of therapeutic strategy for PE by targeting Nesfatin-1.


Assuntos
Movimento Celular , Proliferação de Células , Glicogênio Sintase Quinase 3 beta/metabolismo , Nucleobindinas/metabolismo , Estresse Oxidativo , Fosfatidilinositol 3-Quinase/metabolismo , Pré-Eclâmpsia/enzimologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Trofoblastos/enzimologia , Linhagem Celular , Ativação Enzimática , Transição Epitelial-Mesenquimal , Feminino , Pontos de Checagem da Fase G1 do Ciclo Celular , Humanos , Nucleobindinas/genética , Pré-Eclâmpsia/genética , Pré-Eclâmpsia/patologia , Gravidez , Transdução de Sinais , Trofoblastos/patologia
8.
Reprod Sci ; 28(1): 178-185, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32676926

RESUMO

Preeclampsia is a multi-system disease that is unique to human pregnancy. Impaired extravillous trophoblast migration and invasion accompanied by poor spiral vascular remodeling is thought to be the initial reason. This study investigated cAMP-dependent protein kinase inhibitor-b(PKIB) expression in placentas and its involvement in the pathogenesis of PE. We used immunohistochemistry and western blotting to calculate PKIB levels in the placentas. Then we knocked down PKIB by siRNA and used real-time cell analysis to assess the invasion and migration ability of trophoblasts. Tube formation assay and spheroid sprouting assay were utilized to identify the ability to form vessels of trophoblasts. At last, western blotting was used to demonstrate the level of phosphorylated Akt, as well as downstream-related genes of Akt signaling pathway in trophoblasts. We first found that PKIB expression level was lower in the PE placentas than in the normal placentas. In addition, we found that downregulation of PKIB can inhibit the migration, invasion, and the ability to form vessels of HTR8/SVneo cells. Downregulation of PKIB leaded to a decrease in phosphorylated Akt, as well as downstream proteins such as matrix metalloproteinase 2, matrix metalloproteinase 9, and glycogen synthase kinase 3ß, which are related to migration and invasion. Our study revealed that the downregulation of PKIB expression resulted in decreased migration, invasion, and vessel formation ability by regulating Akt signaling pathway in placental trophoblasts in PE.


Assuntos
Movimento Celular , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Pré-Eclâmpsia/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Trofoblastos/enzimologia , Adulto , Estudos de Casos e Controles , Linhagem Celular , Regulação para Baixo , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Fosforilação , Pré-Eclâmpsia/diagnóstico , Pré-Eclâmpsia/fisiopatologia , Gravidez , Transdução de Sinais , Trofoblastos/patologia , Remodelação Vascular
9.
Cell Biol Int ; 45(3): 599-611, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33200474

RESUMO

Placental hypoxia has been implicated in pregnancy pathologies such as pre-eclampsia and intrauterine growth restriction. However, the underlying mechanism by which the trophoblasts respond to hypoxia remains unclear. Speckle-type POZ protein (SPOP), an E3 ubiquitin ligase adapter, was previously reported to play important roles in various physiological and pathological processes. This study aims to investigate the expression and biological functions of SPOP after exposure to cobalt chloride (CoCl2 )-mimicked hypoxia conditions using human trophoblast-derived choriocarcinoma cell lines and extravillous cytotrophoblast. These data showed that SPOP protein was directly induced by CoCl2 -mimicked hypoxia and regulated by HIF-1α at the posttranscription level. CoCl2 treatment could dramatically influence the localization of SPOP in trophoblasts, especially the accumulation of SPOP into the nucleus. In addition, both CoCl2 -mimicked hypoxia and induction of endogenous SPOP expression by lentivirus transfection attenuated the migration and invasion abilities of trophoblasts. Furthermore, we demonstrated that SPOP was involved in CoCl2 -induced the inhibition of the PI3K/AKT/GSK3ß pathway in placental trophoblasts. Taken together, these data indicate that accumulation of HIF-1α augments the expression of SPOP in trophoblasts, which impairs trophoblastic mobility by targeting the PI3K/AKT/GSK3ß pathway. This potentially leads to insufficient uterine spiral artery remodeling and suboptimal placental perfusion, and thus the development of pregnancy-related complication.


Assuntos
Movimento Celular , Glicogênio Sintase Quinase 3 beta/metabolismo , Proteínas Nucleares/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais , Trofoblastos/enzimologia , Trofoblastos/patologia , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Cobalto/toxicidade , Feminino , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Placenta/patologia , Gravidez , Estabilidade Proteica/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Trofoblastos/efeitos dos fármacos
10.
mBio ; 11(5)2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32934082

RESUMO

Zika virus (ZIKV) infection during pregnancy causes intrauterine growth defects and microcephaly, but knowledge of the mechanism through which ZIKV infects and replicates in the placenta remains elusive. Here, we found that ALPP, an alkaline phosphatase expressed primarily in placental tissue, promoted ZIKV infection in both human placental trophoblasts and astrocytoma cells. ALPP bound to ZIKV structural and nonstructural proteins and thereby prevented their proteasome-mediated degradation and enhanced viral RNA replication and virion biogenesis. In addition, the function of ALPP in ZIKV infection depends on its phosphatase activity. Furthermore, we demonstrated that ALPP was stabilized through interactions with BIP, which is the endoplasmic reticulum (ER)-resident heat shock protein 70 chaperone. The chaperone activity of BIP promoted ZIKV infection and mediated the interaction between ALPP and ZIKV proteins. Collectively, our findings reveal a previously unrecognized mechanism through which ALPP facilitates ZIKV replication by coordinating with the BIP protein.IMPORTANCE ZIKV is a recently emerged mosquito-borne flavivirus that can cause devastating congenital Zika syndrome in pregnant women and Guillain-Barré syndrome in adults, but how ZIKV specifically targets the placenta is not well understood. Here, we identified an alkaline phosphatase (ALPP) that is expressed primarily in placental tissue and promotes ZIKV infection by colocalizing with ZIKV proteins and preventing their proteasome-mediated degradation. The phosphatase activity of ALPP could be required for optimal ZIKV infection, and ALPP is stabilized by BIP via its chaperone activity. This report provides novel insights into host factors required for ZIKV infection, which potentially has implications for ZIKV infection of the placenta.


Assuntos
Fosfatase Alcalina/genética , Fosfatase Alcalina/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Placenta/virologia , Proteínas Virais/metabolismo , Replicação Viral/genética , Zika virus/fisiologia , Aedes , Animais , Astrocitoma , Linhagem Celular , Feminino , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Células HEK293 , Humanos , Placenta/citologia , Placenta/enzimologia , Gravidez , Trofoblastos/enzimologia , Trofoblastos/virologia , Replicação Viral/fisiologia , Zika virus/genética , Infecção por Zika virus
11.
Biosci Rep ; 40(6)2020 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-32342983

RESUMO

Preeclampsia (PE), a common obstetrical disorder, is characterized by impaired migration and invasion abilities of trophoblastic cells. MicroRNA-183-5p (miR-183) was reported to regulate cell migration and invasion in various types of human cancers; however, its role in the pathogenesis of PE remains elusive. Herein, we investigated the role of miR-183 in HTR-8/SVneo trophoblast cells invasion and migration and explored the underlying mechanism. Our results showed that miR-183 was significantly up-regulated in placental tissues from pregnant women compared with that in normal pregnant women. Overexpression of miR-183 inhibited proliferation, migration and invasion, as well as induced apoptosis in HTR-8/SVneo cells. Otherwise, down-regulation of miR-183 achieved the opposite effects. Bioinformatics prediction and luciferase reporter assay confirmed that matrix metalloproteinase-9 (MMP-9) is a target of miR-183. In addition, MMP-9 expression was significantly down-regulated, and inversely correlated with the miR-183 level in placental tissues from pregnant women with severe PE. Down-regulation of MMP-9 suppressed the trophoblast cell invasion and migration, whereas overexpression of MMP-9 promoted cell invasion and migration in HTR-8/SVneo cells. More importantly, up-regulation of MMP-9 reversed the inhibitory effects of miR-183 on cell invasion and migration in trophoblast cells. Collectively, our findings suggested that miR-183 may play critical roles in the pathogenesis of PE and serve as a potential biomarker for severe PE.


Assuntos
Movimento Celular , Metaloproteinase 9 da Matriz/metabolismo , MicroRNAs/metabolismo , Pré-Eclâmpsia/enzimologia , Trofoblastos/enzimologia , Adulto , Apoptose , Estudos de Casos e Controles , Linhagem Celular , Proliferação de Células , Feminino , Regulação Enzimológica da Expressão Gênica , Humanos , Metaloproteinase 9 da Matriz/genética , MicroRNAs/genética , Pré-Eclâmpsia/genética , Pré-Eclâmpsia/patologia , Gravidez , Transdução de Sinais , Trofoblastos/patologia , Adulto Jovem
12.
J Recept Signal Transduct Res ; 40(3): 218-223, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32069432

RESUMO

Purpose: Excessive inflammatory response is one of the possible pathogenic mechanisms of preeclampsia (PE). It remains unclear whether sevoflurane has an anti-inflammatory effect in human trophoblastic cells, which are corresponding to the dysfunction of placentas in PE. This study probed into the regulatory function of sevoflurane toward HTR8/SVneo cells so as to find PE pathology and PE treatment.Materials and methods: HTR8/SVneo cells were treated with sevoflurane, TNF-α with different concentrations, sevoflurane plus 10 ng/mL TNF-α and SB203580 plus 10 ng/mL TNF-α. Cell counting kit-8 (CCK-8) assays were performed to detect cell viability, while enzyme linked immunoSorbent assay (ELISA) was used to measure IL-6, IL-8, GM-CSF and MCP-1 levels in HTR8/SVneo cells. Besides, relative mRNA expression levels of IL-6 and IL-8 were tested via quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR), and p38 phosphorylation-related protein expressions were assessed through western blot.Results: Cell viability remained stable when HTR8/SVneo cells were treated with or without sevoflurane and SB203580 in inflammatory microenvironment created by TNF-α. MCP-1 and GM-CSF levels, as well as gene expressions of IL-6 and IL-8 in HTR8/SVneo cells were greatly increased by TNF-α (5, 10 and 20 ng/mL), but reversed by sevoflurane and SB203580. Simultaneously, TNF-α-induced phosphorylation of p38MAPK signaling pathway was inhibited by sevoflurane and SB203580.Conclusions: Sevoflurane inhibited inflammatory response induced by TNF-α in human trophoblastic cells HTR8/SVneo through suppressing the phosphorylation of p38MAPK signaling pathway.


Assuntos
Inflamação/patologia , Sistema de Sinalização das MAP Quinases , Sevoflurano/farmacologia , Trofoblastos/enzimologia , Trofoblastos/patologia , Fator de Necrose Tumoral alfa/efeitos adversos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Humanos , Imidazóis/farmacologia , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Piridinas/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Trofoblastos/efeitos dos fármacos
13.
Biosci Rep ; 40(2)2020 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-32039444

RESUMO

Despite years of study, the gestational disorder preeclampsia (PE) remains poorly understood. One proposed mechanism of PE development is increased soluble VEGF receptor-1 (sFlt-1), ultimately causing angiogenic imbalance and endothelial dysfunction. The soluble protein is an alternative splice variant of FLT1, which also encodes for the full-length receptor Flt-1. The mechanism of the alternative splicing, and the reason for its inappropriate increase in preeclampsia, is not well understood. U2 auxiliary factor 65 (U2AF65) and jumonji C domain-containing protein 6 (JMJD6) have been implicated in the splicing of sFlt-1. Using siRNA knockdown and plasmid overexpression in immortalized placental trophoblasts (BeWo) and primary endothelial cells (HUVECs), we examined the role these proteins play in production of sFlt-1. Our results showed that U2AF65 has little, if any, effect on sFlt-1 splicing, and JMJD6 may enhance sFlt-1 splicing, but is not necessary for splicing to occur. Utilizing a hypoxic environment to mimic conditions of the preeclamptic placenta, as well as examining placentae in the reduced uterine perfusion pressure (RUPP) model of PE, which exhibits increased circulating sFlt-1, we found increased expression of JMJD6 in both hypoxic cells and placental tissue. Additionally, we observed a potential role for U2AF65 and JMJD6 to regulate the extracellular matrix enzyme heparanase, which may be involved in the release of sFlt-1 protein from the extracellular matrix. It will be important to study the role of these proteins in different tissues in the future, as changes in expression had differential effects on sFlt-1 splicing in the different cell types studied here.


Assuntos
Processamento Alternativo , Células Endoteliais da Veia Umbilical Humana/enzimologia , Histona Desmetilases com o Domínio Jumonji/metabolismo , Pré-Eclâmpsia/enzimologia , Fator de Processamento U2AF/metabolismo , Trofoblastos/enzimologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Hipóxia Celular , Linhagem Celular , Modelos Animais de Doenças , Feminino , Glucuronidase/metabolismo , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Pré-Eclâmpsia/genética , Pré-Eclâmpsia/fisiopatologia , Gravidez , Ratos Sprague-Dawley , Fator de Processamento U2AF/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética
14.
J Mol Med (Berl) ; 98(1): 123-133, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31832700

RESUMO

Preeclampsia (PE) is the most threatening pathology of human pregnancy. Its development is thought to be due to a failure in the invasion of trophoblast cells that establish the feto-maternal circulation. Protein kinase CK2 is a ubiquitous enzyme reported to be involved in the control of cell invasion. CK2 consists of two subunits, a catalytic subunit, CK2α, and a regulatory subunit, CK2ß. To date, no data exist regarding the expression and role of this enzyme in normal and PE pregnancies. We performed studies, at the clinical level using distinctive cohorts from early pregnancy (n = 24) and from PE (n = 23) and age-matched controls (n = 28); in vitro, using trophoblast cell lines; ex vivo, using placental explants; and in vivo, using PE mouse models. We demonstrated that (i) CK2 is more expressed during the late first trimester of pregnancy and is mainly localized in differentiated trophoblast cells, (ii) the inhibition of its enzymatic activity decreased the proliferation, migration, invasion, and syncytialization of trophoblast cells, both in 2D and 3D culture systems, and (iii) CK2 activity and the CK2α/CK2ß protein ratio were increased in PE human placentas. The pattern and profile of CK2 expression were confirmed in gravid mice along with an increase in the PE mouse models. Altogether, our results demonstrate that CK2 plays an essential role in the establishment of the feto-maternal circulation and that its deregulation is associated with PE development. The increase in CK2 activity in PE might constitute a compensatory mechanism to ensure proper pregnancy progress.


Assuntos
Placenta/enzimologia , Placentação , Pré-Eclâmpsia/enzimologia , Trofoblastos/enzimologia , Adolescente , Adulto , Animais , Caseína Quinase II/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Placenta/patologia , Pré-Eclâmpsia/patologia , Gravidez , Primeiro Trimestre da Gravidez , Adulto Jovem
15.
Theranostics ; 9(13): 3853-3865, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31281518

RESUMO

N6-Methyladenosine (m6A) is the most prevalent internal modification in mammalian mRNAs. Although m6A is important in many biological processes, its roles in the placenta are unclear. Methods: Levels of global mRNA m6A methylation and ALKBH5 expression in recurrent miscarriage (RM) patients were determined using quantitative reverse transcription-PCR (qRT-PCR), m6A RNA methylation quantification, and immunohistochemical methods. Using ALKBH5 overexpression and knockdown methods, we determined the role of ALKBH5 in trophoblast invasion at the maternal interface through trophoblasts and an extravillous explant culture experiments. Furthermore, the regulation of CYR61 by ALKBH5 was explored by RNA-sequencing coupled with methylated RNA immunoprecipitation. Results: We found that the level of global mRNA m6A methylation was significantly decreased in placental villous tissue from RM patients, while ALKBH5 expression was specifically unregulated. Furthermore, we demonstrated that ALKBH5 knockdown in human trophoblast promoted trophoblast invasion. Conversely, overexpression of ALKBH5 inhibited cell invasion. ALKBH5 knockdown promoted trophoblast invasion in villous explant culture experiments, while overexpression of ALKBH5 repressed these effects. Furthermore, we clarified that ALKBH5 inhibited trophoblast invasion by regulating CYR61 mRNA stability, and this RNA regulation is m6A dependent. Mechanistic analyses showed that decreased ALKBH5 in trophoblast increased the half-life of CYR61 mRNA and promoted steady-state CYR61 mRNA expression levels. Conclusions: We elucidated the functional roles of ALKBH5 and mRNA m6A methylation in trophoblast and identified a novel RNA regulatory mechanism, providing a basis for further exploration of broad RNA epigenetic regulatory patterns in RM diseases.


Assuntos
Adenosina/análogos & derivados , Homólogo AlkB 5 da RNA Desmetilase/metabolismo , Proteína Rica em Cisteína 61/genética , Troca Materno-Fetal/genética , Estabilidade de RNA/genética , Trofoblastos/citologia , Trofoblastos/enzimologia , Aborto Habitual/genética , Adenosina/metabolismo , Adulto , Movimento Celular/genética , Proteína Rica em Cisteína 61/metabolismo , Feminino , Humanos , Metilação , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Regulação para Cima/genética , Adulto Jovem
16.
Cell Microbiol ; 21(10): e13080, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31265755

RESUMO

Brucella melitensis infection causes acute necrotizing inflammation in pregnant animals; however, the pathophysiological mechanisms leading to placentitis are unknown. Here, we demonstrate that high-mobility group box 1 (HMGB1) acts as a mediator of placenta inflammation in B. melitensis-infected pregnant mice model. HMGB1 levels were increased in trophoblasts or placental explant during B. melitensis infection. Inhibition of HMGB1 activity with neutralising antibody significantly reduced the secretion of inflammatory cytokines in B. melitensis-infected trophoblasts or placenta, whereas administration of recombinant HMGB1 (rHMGB1) increased the inflammatory response. Mechanistically, this decreased inflammatory response results from inhibition of HMGB1 activity, which cause the suppression of both mitogen-activated protein kinases and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activation. Moreover, neutralising antibody to HMGB1 prevented B. melitensis infection-induced activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase in trophoblasts. In contrast, in vitro stimulation of trophoblasts with rHMGB1 caused activation of NADPH oxidase and increased the production of ROS, which contributes to high bacterial burden within trophoblasts or placenta. In vivo, treatment with anti-HMGB1 antibody increases the number of Brucella survival within placenta in B. melitensis-infected pregnant mice but successfully reduced the severity of placentitis and abortion.


Assuntos
Brucella melitensis/fisiologia , Brucelose/imunologia , Proteína HMGB1/metabolismo , Placenta/imunologia , Trofoblastos/metabolismo , Trofoblastos/microbiologia , Aborto Espontâneo/genética , Aborto Espontâneo/metabolismo , Aborto Espontâneo/microbiologia , Animais , Brucella melitensis/genética , Brucella melitensis/metabolismo , Brucella melitensis/patogenicidade , Brucelose/genética , Brucelose/metabolismo , Citocinas/metabolismo , Replicação do DNA/imunologia , Feminino , Proteína HMGB1/administração & dosagem , Proteína HMGB1/antagonistas & inibidores , Proteína HMGB1/genética , Inflamação/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NADPH Oxidases/química , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Fosforilação , Placenta/microbiologia , Placenta/patologia , Gravidez , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Trofoblastos/enzimologia
17.
J Immunol ; 203(5): 1198-1207, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31315888

RESUMO

It is increasingly recognized that excessive glucocorticoids induce fetal intrauterine growth restriction (IUGR). Placental 11ß-hydroxysteroid dehydrogenase 2 (11ß-HSD2), a glucocorticoid-catalyzing enzyme, prevents active glucocorticoids from maternal circulation into the fetus, thus protecting against IUGR. Previous studies demonstrated gestational LPS exposure caused fetal IUGR. The aim of the current study was to investigate the effects of LPS on 11ß-HSD2 in mice placentas and human placental trophoblasts. Pregnant ICR(CD-1) mice were i.p. injected with LPS (200 µg/kg) on gestational day 16. As expected, gestational LPS exposure downregulated 11ß-HSD2 in mice placentas. In vitro, LPS downregulated 11ß-HSD2 in human placental trophoblasts. Additional experiment showed that LPS, which activated NF-κB, suppressed rosiglitazone-induced activation of peroxisome proliferator-activated receptor-γ (PPARγ) in mice placentas and human placental trophoblasts. Moreover, NF-κB p65 knockdown and specific NF-κB inhibitor attenuated LPS-induced suppression of PPARγ nuclear translocation in human placental trophoblasts. In addition, NF-κB p65 knockdown attenuated LPS-induced downregulation of 11ß-HSD2 in human placental trophoblasts. Mechanically, LPS promoted physical interaction between NF-κB p65 and PPARγ in the cytoplasm and nucleus of placental trophoblasts. Finally, pretreatment with rosiglitazone, a PPARγ agonist, partially alleviated LPS-induced reduction of fetal weight and crown-rump length. Taken together, these results suggest that LPS downregulates 11ß-HSD2 through suppressing PPARγ in placental trophoblasts. Placental 11ß-HSD2 downregulation may contribute partially to LPS-induced fetal IUGR.


Assuntos
11-beta-Hidroxiesteroide Desidrogenases/genética , Lipopolissacarídeos/toxicidade , PPAR gama/antagonistas & inibidores , Placenta/efeitos dos fármacos , Trofoblastos/efeitos dos fármacos , Transporte Ativo do Núcleo Celular , Animais , Células Cultivadas , Regulação para Baixo , Feminino , Retardo do Crescimento Fetal/induzido quimicamente , Humanos , Masculino , Camundongos , Camundongos Endogâmicos ICR , PPAR gama/fisiologia , Placenta/enzimologia , Gravidez , Rosiglitazona/farmacologia , Fator de Transcrição RelA/antagonistas & inibidores , Fator de Transcrição RelA/fisiologia , Trofoblastos/enzimologia
18.
Reprod Sci ; 26(7): 961-971, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30305007

RESUMO

Preeclampsia (PE) remains the leading cause of maternal and fetal morbidity and mortality. Excessive apoptosis of the placenta and poor remodeling of spiral arteries caused by insufficient invasion of trophoblast cells into uterus have been implicated in the pathogenesis of PE. Accumulating evidence showed that heat shock protein 20 (HSP20) is closely associated with the proliferation, apoptosis, and metastasis of tumor cells. However, little is known about whether HSP20 plays a role in the development of PE. In this study, we detected the apoptosis index and the expressions of HSP20 and apoptosis-associated proteins in the placentas from PE and normal pregnancies. We found that HSP20 was reversely related to the apoptosis rate and the levels of proapoptotic proteins. Moreover, we identified that HSP20 could suppress the proliferation and apoptosis of trophoblast cells, turning them into a more invasive phenotype. Additionally, H2O2-induced oxidative stress was significantly alleviated, and several key proteins on the Akt signaling pathway were upregulated in HSP20-overexpressing trophoblast cells. These findings strongly suggested that HSP20 might play a role in the remodeling of spiral arteries through affecting the invasiveness of extravillous trophoblast cells via Akt signaling pathway, and the dysregulation of it might contribute to the pathophysiology of PE.


Assuntos
Proteínas de Choque Térmico HSP20/metabolismo , Pré-Eclâmpsia/enzimologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Trofoblastos/enzimologia , Adulto , Apoptose , Proteínas Reguladoras de Apoptose/metabolismo , Estudos de Casos e Controles , Linhagem Celular , Movimento Celular , Proliferação de Células , Feminino , Proteínas de Choque Térmico HSP20/genética , Humanos , Estresse Oxidativo , Pré-Eclâmpsia/genética , Pré-Eclâmpsia/patologia , Gravidez , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Trofoblastos/patologia
19.
Clin Sci (Lond) ; 132(20): 2221-2239, 2018 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-30287519

RESUMO

Preterm labor is associated with inflammation and infection. The mechanisms underlying the role of omega-3 fatty acid in inflammasome activation and prevention of preterm labor remain unknown. We hypothesized that omega-3 fatty acid can reduce the rate of preterm birth induced by infection and trophoblast inflammation. In the present study, we found that inflammasome-related molecules and IL-1ß in trophoblasts were activated by TNF-α derived from lipopolysaccharide (LPS)-stimulated THP-1 cell-conditioned medium (CM) and recombinant TNF-α protein. The results demonstrated that stimulation with TNF-α caused lysosomal rupture in trophoblasts, which accelerated cathepsin S (CTSS) diffusion from lysosomes to the cytosol and activated NLRP1 (nacht domain-leucine-rich repeat, and pyd-containing protein 1) and absent in melanoma 2 (AIM2) inflammasomes, thereby increasing IL-1ß secretion. Moreover, in response to LPS challenge, TNF-α increased trophoblast cell death and decreased cell viability through inflammasome and CTSS activation. Stearidonic acid (SDA; 18:4n-3) and docosahexaenoic acid (DHA; 22:6n-3) inhibited inflammasome-related molecule synthesis and CTSS and caspase-1 activation, which further reduced the preterm delivery rate of pregnant mice induced by LPS (92.9 compared with 69.7% (DHA); 92.9 compared with 53.5% (SDA)). Higher expression of TNF-α, IL-1ß, prostaglandin E2, and CTSS, but lower resolvin D1 expression, was observed in preterm pregnant mice than in controls. Similarly, resolvin D1 was highly expressed in women with term delivery compared with women with preterm delivery. Thus, SDA and DHA may attenuate macrophage-derived TNF-α inducing CTSS and inflammasome activation, IL-1ß secretion, and placental trophoblast cell death. These functions are implicated in the preventive effects of SDA and DHA on preterm labor.


Assuntos
Catepsinas/antagonistas & inibidores , Ácidos Graxos Ômega-3/farmacologia , Inflamassomos/efeitos dos fármacos , Trabalho de Parto Prematuro/prevenção & controle , Trofoblastos/enzimologia , Animais , Catepsinas/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Ácidos Docosa-Hexaenoicos/farmacologia , Feminino , Células HEK293 , Humanos , Inflamassomos/metabolismo , Interleucina-1beta/metabolismo , Lipopolissacarídeos/farmacologia , Camundongos , Gravidez , Células THP-1
20.
Eur Rev Med Pharmacol Sci ; 22(19): 6190-6197, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30338781

RESUMO

OBJECTIVE: To detect the differentially expressed long non-coding RNA (lncRNA) EGFR-AS1 in the placenta tissues of preeclampsia (PE) patients and normal pregnancies. We also investigated the underlying mechanism of EGFR-AS1 in regulating PE development. PATIENTS AND METHODS: 21 PE patients and 21 normal pregnancies admitted in our hospital were selected. The expression of EGFR Antisense RNA1 (EGFR-AS1) in the placenta tissues of PE patients and normal pregnancies was detected by quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR). The regulatory effect of EGFR-AS1 on proliferation of HTR-8 cells was observed by Cell Counting Kit-8 (CCK-8) assay and colony formation assay. Cell cycle and apoptosis of HTR-8 cells after overexpression or knockdown of EGFR-AS1 were detected by flow cytometry. The expression changes of EGFR and proteins related to JAK/STAT signaling pathway after knockdown of EGFR-AS1 in HTR-8 cells were detected by Western blot. Rescue experiments were carried out after upregulating EGFR expression in HTR-8 cells. RESULTS: QRT-PCR results showed that the mRNA expression of EGFR-AS1 in PE patients was lower than that in normal pregnancies. CCK-8 assay and colony formation assay showed that knockdown of EGFR-AS1 in HTR-8 cells inhibited cell proliferation. Flow cytometry results showed that knockdown of EGFR-AS1 blocked the cell cycle of HTR-8 cells. Overexpression of EGFR-AS1 obtained opposite results. Western blot results showed that the protein expressions of EGFR, p-JAK and p-STAT were decreased after knockdown of EGFR-AS1. Alterations in protein expressions of p-JAK and p-STAT, and proliferative ability of HTR-8 cells induced by EGFR-AS1 knockdown could be rescued after upregulation of EGFR. CONCLUSIONS: The expression of lncRNA EGFR-AS1 is decreased in PE patients, which can promote the progression of preeclampsia by inhibiting EGFR-JAK/STAT signaling pathway.


Assuntos
Proliferação de Células , Janus Quinases/metabolismo , Pré-Eclâmpsia/enzimologia , RNA Longo não Codificante/metabolismo , Fatores de Transcrição STAT/metabolismo , Trofoblastos/enzimologia , Adulto , Estudos de Casos e Controles , Ciclo Celular , Linhagem Celular , Progressão da Doença , Regulação para Baixo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Feminino , Humanos , Fosforilação , Pré-Eclâmpsia/genética , Pré-Eclâmpsia/patologia , Gravidez , RNA Longo não Codificante/genética , Transdução de Sinais , Trofoblastos/patologia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA