Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Front Endocrinol (Lausanne) ; 14: 1021640, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36936174

RESUMO

Placenta accreta spectrum (PAS) is one of the major causes of maternal morbidity and mortality worldwide with increasing incidence. PAS refers to a group of pathological conditions ranging from the abnormal attachment of the placenta to the uterus wall to its perforation and, in extreme cases, invasion into surrounding organs. Among them, placenta accreta is characterized by a direct adhesion of the villi to the myometrium without invasion and remains the most common diagnosis of PAS. Here, we identify the potential regulatory miRNA and target networks contributing to placenta accreta development. Using small RNA-Seq followed by RT-PCR confirmation, altered miRNA expression, including that of members of placenta-specific miRNA clusters (e.g., C19MC and C14MC), was identified in placenta accreta samples compared to normal placental tissues. In situ hybridization (ISH) revealed expression of altered miRNAs mostly in trophoblast but also in endothelial cells and this profile was similar among all evaluated degrees of PAS. Kyoto encyclopedia of genes and genomes (KEGG) analyses showed enriched pathways dysregulated in PAS associated with cell cycle regulation, inflammation, and invasion. mRNAs of genes associated with cell cycle and inflammation were downregulated in PAS. At the protein level, NF-κB was upregulated while PTEN was downregulated in placenta accreta tissue. The identified miRNAs and their targets are associated with signaling pathways relevant to controlling trophoblast function. Therefore, this study provides miRNA:mRNA associations that could be useful for understanding PAS onset and progression.


Assuntos
MicroRNAs , Placenta Acreta , Gravidez , Humanos , Feminino , Placenta Acreta/genética , Placenta Acreta/metabolismo , Placenta Acreta/patologia , MicroRNAs/genética , MicroRNAs/metabolismo , Células Endoteliais/metabolismo , Placenta/metabolismo , Miométrio
2.
Front Physiol ; 13: 882544, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35707005

RESUMO

The NO-donor Pentaerytrithyltetranitrate (PETN) has vasodilatative properties and direct protective effects on endothelial cells. We formerly demonstrated that PETN, given to pregnant women during the second and third trimester, influences endothelial dysfunction related pregnancy complications like preeclampsia (PE) and fetal growth restriction (FGR). PETN treatment showed to delay PE to late pregnancy and achieved a profound risk reduction for FGR and/or perinatal death of 40%. The aim of this study was to confirm the effect of PETN on endothelial cell dysfunction at molecular level in an experimental approach. To induce endothelial dysfunction HUVEC were treated with 10 U/l of thrombin in the presence or absence of PETN. qRT-PCR analysis showed that PETN induced the expression of heme-oxygenase-1 and superoxide dismutase two but not endothelial NO-synthase under basal conditions. The induction of antioxidant proteins did not change basal reactive oxygen species (ROS) levels as measured by MitoSOX™ staining. PETN treatment significantly delayed the thrombin-induced disruption of the endothelial monolayer, determined using the xCELLigence® and attenuated the disrupting effect of thrombin on tubular junctions as seen in a tube-forming assay on Matrigel™. In western-blot-analysis we could show that PETN significantly reduced thrombin-induced extracellular signal-regulated kinase activation which correlates with reduction of thrombin-induced ROS. These experimental results establish the concept of how PETN treatment could stabilize endothelial resistance and angiogenic properties in pregnancy-induced stress. Thus, our results underscore the assumption, that the shown clinical effects of PETN are associated to its endothelial cell protection.

3.
Mol Aspects Med ; 87: 101023, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-34521556

RESUMO

Tobacco smoking is an important public health issue recognized by the world health organization as one of the most serious, preventable risk factors for developing a series of pregnancy pathologies. Maternal smoking is positively associated with intrauterine growth restriction (IUGR) and gestational diabetes (GDM), but negatively associated with preeclampsia (PE). In this review, we examine epidemiological, clinical and laboratory studies of smoking effects on immunoregulation during pregnancy, trophoblast function, and placental vasculature development and metabolism. We aim to identify effects of tobacco smoke components on specific placental compartments or cells, which may contribute to the understanding of the influences of maternal smoking on placenta function in normal and pathological pregnancies. Data corroborates that in any trimester, smoking is unsafe for pregnancy and that its detrimental effects outweigh questionable benefits. The effects of maternal smoking on the maternal immune regulation throughout pregnancy and the impact of different tobacco products on fetal growth have not yet been fully understood. Smoking cessation rather than treatment with replacement therapies is recommended for future mothers because also single components of tobacco and its smoke may have detrimental effects on placental function.


Assuntos
Placenta , Fumar , Feminino , Retardo do Crescimento Fetal/epidemiologia , Retardo do Crescimento Fetal/etiologia , Retardo do Crescimento Fetal/metabolismo , Humanos , Placenta/metabolismo , Gravidez , Fumar/efeitos adversos , Fumar/metabolismo , Fumar Tabaco , Uso de Tabaco , Trofoblastos/metabolismo
4.
Biochim Biophys Acta Mol Basis Dis ; 1867(11): 166218, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34311080

RESUMO

Throughout history, pandemics of infectious diseases caused by emerging viruses have spread worldwide. Evidence from previous outbreaks demonstrated that pregnant women are at high risk of contracting the diseases and suffering from adverse outcomes. However, while some viruses can cause major health complications for the mother and her fetus, others do not appear to affect pregnancy. Viral surface proteins bind to specific receptors on the cellular membrane of host cells and begin therewith the infection process. During pregnancy, the molecular features of these proteins may determine specific target cells in the placenta, which may explain the different outcomes. In this review, we display information on Variola, Influenza, Zika and Corona viruses focused on their surface proteins, effects on pregnancy, and possible target placental cells. This will contribute to understanding viral entry during pregnancy, as well as to develop strategies to decrease the incidence of obstetrical problems in current and future infections.


Assuntos
Placenta/virologia , Complicações Infecciosas na Gravidez/virologia , Proteínas do Envelope Viral/metabolismo , Viroses/virologia , Feminino , Humanos , Placenta/metabolismo , Gravidez , Complicações Infecciosas na Gravidez/metabolismo , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidade , Vírus da Varíola/metabolismo , Vírus da Varíola/patogenicidade , Viroses/metabolismo , Zika virus/metabolismo , Zika virus/patogenicidade
5.
Placenta ; 108: 122-133, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33810901

RESUMO

INTRODUCTION: Research on human placental development and function lacks a conclusive in vivo model. To investigate the intracellular molecular mechanisms in trophoblast cells, different cell lines have been established during the last decades. So far, none of these accomplishes all features of primary trophoblast, thus their suitability as well as the transferability of the results has been discussed. The aim of this study is to assess molecular markers and features matching different trophoblast subpopulations in trophoblastic cell lines to provide orientation on their suitability and relevance for distinct research questions. METHODS: The commonly used trophoblastic cell lines, BeWo, JEG-3, HTR-8/SVneo, AC1-M59, AC1-M32, ACH-3P and Swan71 were selected. qPCR and immunoblotting were used to determine expression of characteristic molecular markers. C14MC, C19MC and miR-371-3 miRNA expression were investigated by real time PCR. Proliferation, migration and network stabilization assays were performed. Hormone secretion was determined by chemiluminescent-immunoassays. DNA profiles were obtained by Short Tandem Repeat (STR)-genotyping. RESULTS: Immortalized cell lines differ from choriocarcinoma-derived ones in the expression of HLA-G, E-cadherin, N-cadherin, VE-cadherin, cadherin-11, cytokeratin 7, vimentin, ADAM12 and PRG2. Compared to choriocarcinoma-derived cell lines, expression of C19MC and hormone secretion were almost absent in immortalized cell lines. Conversely, they express C14MC and exhibit higher migration and network stabilization. DISCUSSION: The data presented will help justify the use of a cell line to evaluate distinct features of trophoblast biology and pathology. In general, characteristics and markers of choriocarcinoma derived cell lines seem to be more similar to in vivo trophoblast than immortalized cell lines and thus might be regarded as more suitable models.


Assuntos
MicroRNAs/metabolismo , Placenta/metabolismo , Trofoblastos/metabolismo , Animais , Linhagem Celular , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Feminino , Humanos , Gravidez
6.
Am J Reprod Immunol ; 85(2): e13383, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33251688

RESUMO

Extracellular vesicle (EV)-mediated communication has been implicated in the cooperative alliance between trophoblast and immune cells toward maternal tolerance and placentation. Syncytiotrophoblast cells secrete EVs directly into the maternal circulation, which are taken up by immune cells, endothelial cells, and other cell types. Initial evidence also shows that EVs produced by immune cells are, in turn, incorporated by trophoblast cells and modulate placental responses. Non-coding RNAs (ncRNAs), proteins, and lipid mediators transported in EVs are able to influence proliferation, differentiation, cytokine production, and immunological responses of recipient cells. The molecular alphabet and cellular targets involved in this dialogue are being revealed. Nevertheless, several questions regarding the whole content, surface markers, and biological functions of EVs still remain to be investigated in both physiological and pathological conditions. Analysis of circulating EVs in maternal blood has the potential to serve as a minimally invasive approach to monitoring placental functions and immunological features of pregnancy, aiding in the diagnostics of complications. This review addresses the immunomodulatory properties of EVs and their tasks in the communication between placental and immune cells.


Assuntos
Vesículas Extracelulares/imunologia , Placenta/imunologia , Gravidez/imunologia , Trofoblastos/imunologia , Animais , Biomarcadores , Comunicação Celular , Feminino , Humanos , Tolerância Imunológica , Imunidade Celular , Imunomodulação , Placentação
7.
Int J Mol Sci ; 21(10)2020 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-32422900

RESUMO

Members of the placenta-specific miRNA cluster C19MC, including miR-519d, are secreted by fetal trophoblast cells within extracellular vesicles (EVs). Trophoblast-derived EVs can be internalized by the autologous trophoblast and surrounding maternal immune cells, resulting in coordination of cellular responses. The study of functions and targets of placental miRNAs in the donor and recipient cells may contribute to the understanding of the immune tolerance essential in pregnancy. Here, we report that miR-519d-3p levels correlate positively with cell proliferation and negatively with migration in trophoblastic cell lines. Inhibition of miR-519d-3p in JEG-3 cells increases caspase-3 activation and apoptosis. PDCD4 and PTEN are targeted by miR-519d-3p in a cell type-specific manner. Transfection of trophoblastic cell lines with miR-519d mimic results in secretion of EVs containing elevated levels of this miRNA (EVmiR-519d). Autologous cells enhance their proliferation and decrease their migration ability when treated with EVmiR-519d. NK92 cells incorporate EV-delivered miR-519d-3p at higher levels than Jurkat T cells. EVmiR-519d increases the proliferation of Jurkat T cells but decreases that of NK92 cells. Altogether, miR-519d-3p regulates pivotal trophoblast cell functions, can be transferred horizontally via EVs to maternal immune cells and exerts functions therein. Vesicular miRNA transfer from fetal trophoblasts to maternal immune cells may contribute to the immune tolerance in pregnancy.


Assuntos
Proteínas Reguladoras de Apoptose/genética , MicroRNAs/genética , PTEN Fosfo-Hidrolase/genética , Proteínas de Ligação a RNA/genética , Trofoblastos/metabolismo , Apoptose/genética , Caspase 3/genética , Movimento Celular/genética , Proliferação de Células/genética , Vesículas Extracelulares/genética , Vesículas Extracelulares/imunologia , Feminino , Humanos , Tolerância Imunológica/genética , Tolerância Imunológica/imunologia , Células Jurkat , Células Matadoras Naturais/imunologia , Placenta/imunologia , Placenta/metabolismo , Placentação/genética , Gravidez , Linfócitos T/imunologia , Trofoblastos/imunologia
8.
Int J Mol Sci ; 22(1)2020 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-33396613

RESUMO

IL-36 cytokines (the agonists IL-36α, IL-36ß, IL-36γ, and the antagonist IL-36Ra) are expressed in the mouse uterus and associated with maternal immune response during pregnancy. Here, we characterize the expression of IL-36 members in human primary trophoblast cells (PTC) and trophoblastic cell lines (HTR-8/SVneo and JEG-3) and upon treatment with bacterial and viral components. Effects of recombinant IL-36 on the migration capacity of trophoblastic cells, their ability to interact with endothelial cells and the induction of angiogenic factors and miRNAs (angiomiRNAs) were examined. Constitutive protein expression of IL-36 (α, ß, and γ) and their receptor (IL-36R) was found in all cell types. In PTC, transcripts for all IL-36 subtypes were found, whereas in trophoblastic cell lines only for IL36G and IL36RN. A synthetic analog of double-stranded RNA (poly I:C) and lipopolysaccharide (LPS) induced the expression of IL-36 members in a cell-specific and time-dependent manner. In HTR-8/SVneo cells, IL-36 cytokines increased cell migration and their capacity to interact with endothelial cells. VEGFA and PGF mRNA and protein, as well as the angiomiRNAs miR-146a-3p and miR-141-5p were upregulated as IL-36 response in PTC and HTR-8/SVneo cells. In conclusion, IL-36 cytokines are modulated by microbial components and regulate trophoblast migration and interaction with endothelial cells. Therefore, a fundamental role of these cytokines in the placentation process and in response to infections may be expected.


Assuntos
Regulação da Expressão Gênica/genética , Interleucina-1/genética , Neovascularização Fisiológica/genética , Trofoblastos/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular/genética , Movimento Celular/fisiologia , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-1/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Lipopolissacarídeos/farmacologia , MicroRNAs/genética , Neovascularização Fisiológica/fisiologia , Poli I-C/farmacologia , Prostaglandinas F/genética , Prostaglandinas F/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Trofoblastos/citologia , Trofoblastos/fisiologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
9.
Placenta ; 88: 20-27, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31586768

RESUMO

INTRODUCTION: Leukemia Inhibitory Factor (LIF) regulates behavior of trophoblast cells and their interaction with immune and endothelial cells. In vitro, trophoblast cell response to LIF may vary depending on the cell model. Reported differences in the miRNA profile of trophoblastic cells may be responsible for these observations. Therefore, miRNA expression was investigated in four trophoblastic cell lines under LIF stimulation followed by in silico analysis of altered miRNAs and their associated pathways. METHODS: Low density TaqMan miRNA assays were used to quantify levels of 762 mature miRNAs under LIF stimulation in three choriocarcinoma-derived (JEG-3, ACH-3P and AC1-M59) and a trophoblast immortalized (HTR-8/SVneo) cell lines. Expression of selected miRNAs was confirmed in primary trophoblast cells and cell lines by qPCR. Targets and associated pathways of the differentially expressed miRNAs were inferred from the miRTarBase followed by a KEGG Pathway Enrichment Analysis. HTR-8/SVneo and JEG-3 cells were transfected with miR-21-mimics and expression of miR-21 targets was assessed by qPCR. RESULTS: A similar number of miRNAs changed in each tested cell line upon LIF stimulation, however, low coincidence of individual miRNA species was observed and occurred more often among choriocarcinoma-derived cells (complete data set at http://www.ncbi.nlm.nih.gov/geo/ under GEO accession number GSE130489). Altered miRNAs were categorized into pathways involved in human diseases, cellular processes and signal transduction. Six cascades were identified as significantly enriched, including JAK/STAT and TGFB-SMAD. Upregulation of miR-21-3p was validated in all cell lines and primary cells and STAT3 was confirmed as its target. DISCUSSION: Dissimilar miRNA responses may be involved in differences of LIF effects on trophoblastic cell lines.


Assuntos
Fator Inibidor de Leucemia/fisiologia , MicroRNAs/metabolismo , Trofoblastos/fisiologia , Linhagem Celular , Humanos , Fator de Transcrição STAT3/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA