Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Physiol Heart Circ Physiol ; 318(4): H1008-H1017, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32196359

RESUMO

Circulating levels of placental growth factor (PlGF) are significantly reduced in women who develop preeclampsia. Low molecular weight heparin (LMWH) has been shown to acutely elevate circulating PlGF levels in pregnant women at increased risk of preeclampsia. The objective of the current investigation was to determine the mechanisms by which LMWH mediates the extracellular release of PlGF from endothelial cells. Cultured human aortic endothelial cells (HAECs) and human umbilical vein endothelial cells (HUVECs) were exposed to LMWH; PlGF transcription, translation, mobilization, and secretion were then assessed. LMWH significantly increased the release of PlGF from both HAECs and HUVECs. LMWH treatment promoted a significant increase of PlGF-1 mRNA expression in HAECs, accompanied by the intracellular transport and release of PlGF into the conditioned media. LMWH-mediated release of PlGF from HAECs was not directly mediated by extracellular mobilization, synthesis, or stability of PlGF mRNA/protein. LMWH exposure promotes the release of PlGF from endothelial cells through the upregulation of PlGF-1 mRNA expression. Stimulation of circulating PlGF levels by LMWH may be an important mechanism by which LMWH could reduce the risk of preeclampsia or minimize disease severity.NEW & NOTEWORTHY There are few therapeutic options available for the prevention of preeclampsia, a serious hypertensive disorder of pregnancy. Women who subsequently develop preeclampsia exhibit significantly reduced circulating levels of the proangiogenic placental growth factor protein. Low molecular weight heparin (LMWH) has previously been investigated as a preventative therapy against the development of preeclampsia; however, its mechanism of action is not known. The current study determined that LMWH promotes the transcription and release of placental growth factor protein from endothelial cells, providing a mechanistic basis by which LMWH could reduce the risk of preeclampsia or minimize disease severity.


Assuntos
Anticoagulantes/farmacologia , Heparina de Baixo Peso Molecular/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Células Cultivadas , Exocitose , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Proteínas de Membrana/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
2.
Biol Reprod ; 102(5): 1102-1110, 2020 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-31950133

RESUMO

Preeclampsia is a serious hypertensive disorder of pregnancy, which is only cured with delivery of the placenta, thereby commonly necessitating preterm birth of the fetus. Low-molecular-weight heparin (LMWH) has demonstrated potential to reduce the incidence of preeclampsia in high-risk pregnant women, although the underlying mechanism by which LMWH protects against preeclampsia is unknown. Given the complex structure and biologic actions of heparin, we tested the hypothesis that heparin can mediate preeclampsia prevention via nonanticoagulant pathways. We compared the effects of a nonanticoagulant, glycol-split LMWH (gsHep)-rendered nonanticoagulant through disruption of the antithrombin binding regions-with the LMWH dalteparin in the rat reduced uterine perfusion pressure (RUPP) surgical model of preeclampsia. Although RUPP animals exhibit significantly elevated blood pressure and reduced plasma levels of placental growth factor (PGF) compared to sham, neither dalteparin nor gsHep treatment significantly impacted these parameters. However, the observed positive correlation between PGF levels and number of viable fetuses in RUPP-induced animals suggests that reduced PGF levels were predominately due to placental loss. Daily subcutaneous injections of low-dose dalteparin but not gsHep significantly restored fetal growth that was impaired by RUPP surgery. Placentas from RUPP animals exhibited an abnormal labyrinth structure, characterized by expanded sinusoidal blood spaces, relative to sham-operated animals. Morphometric analysis demonstrated that dalteparin but not gsHep treatment normalized development of the labyrinth in RUPP-exposed conceptuses. These data suggest that the antithrombin-binding regions of LMWH are required to confer its protective effects on fetal growth and placental development.


Assuntos
Dalteparina/farmacologia , Desenvolvimento Fetal/efeitos dos fármacos , Heparina/química , Heparina/metabolismo , Doenças Placentárias/prevenção & controle , Pré-Eclâmpsia/prevenção & controle , Animais , Anticoagulantes/química , Anticoagulantes/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Hipertensão , Fator de Crescimento Placentário/genética , Fator de Crescimento Placentário/metabolismo , Pré-Eclâmpsia/metabolismo , Pré-Eclâmpsia/patologia , Gravidez , Ratos , Ratos Sprague-Dawley
3.
J Physiol ; 596(9): 1587-1600, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29468681

RESUMO

KEY POINTS: The post-translational modification of target proteins by SUMOylation occurs in response to stressful stimuli in a variety of organ systems. Small ubiquitin-like modifier (SUMO) isoforms 1-4 have recently been identified in the human placenta, and are upregulated in the major obstetrical complication of pre-eclampsia. This is the first study to characterize the spatiotemporal distribution of SUMO isoforms and their targets during placental development across gestation and in response to stress induced by pre-eclampsia and chorioamnionitis. Keratins were identified as major targets of placental SUMOylation. The interaction with SUMOs and cytoskeletal filaments provides evidence for SUMOylation possibly contributing to underlying dysfunctional trophoblast turnover, which is a hallmark feature of pre-eclampsia. Further understanding the role of individual SUMO isoforms and SUMOylation underlying placental dysfunction may provide a target for a novel therapeutic candidate as an approach for treating pre-eclampsia complicated with placental pathology. ABSTRACT: SUMOylation is a dynamic, reversible post-translational modification that regulates cellular protein stability and localization. SUMOylation occurs in response to various stressors, including hypoxia and inflammation, features common in the obstetrical condition of pre-eclampsia. SUMO isoforms 1-4 have recently been identified in the human placenta, but less is known about their role in response to pre-eclamptic stress. We hypothesized that SUMOylation components have a unique spatiotemporal distribution during placental development and that their subcellular localization can be further modulated by extra-cellular stressors. Placental SUMO expression was examined across gestation. First-trimester human placental explants and JAR cells were subjected to hypoxia or TNF-α cytokine, and subcellular translocation of SUMOs was monitored. SUMOylation target proteins were elucidated using mass spectrometry and proximity ligation assay. Placental SUMO-1 and SUMO-4 were restricted to villous cytotrophoblast cells in first trimester and syncytium by term, while SUMO-2/3 staining was evenly distributed throughout the trophoblast across gestation. In placental villous explants, oxidative stress induced hyperSUMOylation of SUMO-1 and SUMO-4 in the syncytial cytoplasm, whereas SUMO-2/3 nuclear expression increased. Oxidative stress also upregulated cytoplasmic SUMO-1 and SUMO-4 protein expression (P < 0.05), similar to pre-eclamptic placentas. Keratins were identified as major targets of placental SUMOylation. Oxidative stress increased the cytokeratin-7 to SUMO-1 and SUMO-4 interactions, while inflammatory stress increased its interaction with SUMO-2/3. Overall, SUMOs display a unique spatiotemporal distribution in normal human placental development. Our data indicate SUMOylation in pre-eclampsia, which may impair the stability of cytoskeleton filaments and thus promote trophoblast shedding into the maternal circulation in this condition.


Assuntos
Corioamnionite/fisiopatologia , Inflamação/fisiopatologia , Estresse Oxidativo , Placenta/fisiopatologia , Placentação , Pré-Eclâmpsia/fisiopatologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Feminino , Idade Gestacional , Humanos , Queratina-7/metabolismo , Gravidez , Análise Espaço-Temporal , Frações Subcelulares/metabolismo , Sumoilação , Trofoblastos/metabolismo , Ubiquitina/metabolismo
4.
Biol Reprod ; 99(5): 1082-1090, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-29860275

RESUMO

Low molecular weight heparin (LMWH) is being investigated as a potential preventative therapy against preeclampsia. There is evidence suggesting that LMWH may prevent preeclampsia through anticoagulation-independent mechanisms. In this study, we compared the in vitro placental, endothelial, and anti-inflammatory effects of an LMWH (dalteparin) with a nonanticoagulant, glycol-split heparin derivative (gsHep). In contrast with dalteparin, gsHep did not interact with antithrombin III, possess significant anti-Factor Xa activity, or significantly prolong in vitro plasma clotting time. However, dalteparin and gsHep were otherwise mechanistically similar, both interacting with soluble fms-like tyrosine kinase-1 (sFlt1) and promoting release of the pro-angiogenic protein placental growth factor, but not the antiangiogenic sFlt1, from healthy placental villous explants. Placental explant media pretreated with dalteparin or gsHep significantly stimulated endothelial cell tube formation compared to untreated explants. Lastly, dalteparin and gsHep both significantly suppressed inflammation by inhibiting complement activation and leukocyte adhesion to endothelial cells that were activated using serum from preeclamptic women. Our data suggest that nonanticoagulant heparin derivatives may be utilized as a tool to distinguish the anticoagulation-independent mechanisms of LMWH, and provide insight into the role of anticoagulation in the prevention of preeclampsia.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Glicóis/química , Heparina de Baixo Peso Molecular/farmacologia , Células Endoteliais da Veia Umbilical Humana/patologia , Inflamação/fisiopatologia , Placenta/patologia , Pré-Eclâmpsia/fisiopatologia , Transdução de Sinais/efeitos dos fármacos , Anticoagulantes/farmacologia , Adesão Celular/efeitos dos fármacos , Ativação do Complemento/efeitos dos fármacos , Dalteparina/farmacologia , Fator Xa , Feminino , Heparina de Baixo Peso Molecular/química , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Placenta/efeitos dos fármacos , Gravidez , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/efeitos dos fármacos , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
5.
Mol Cell Proteomics ; 10(12): M111.012526, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21986993

RESUMO

Preeclampsia (PE) adversely impacts ~5% of pregnancies. Despite extensive research, no consistent biomarkers or cures have emerged, suggesting that different molecular mechanisms may cause clinically similar disease. To address this, we undertook a proteomics study with three main goals: (1) to identify a panel of cell surface markers that distinguish the trophoblast and endothelial cells of the placenta in the mouse; (2) to translate this marker set to human via the Human Protein Atlas database; and (3) to utilize the validated human trophoblast markers to identify subgroups of human preeclampsia. To achieve these goals, plasma membrane proteins at the blood tissue interfaces were extracted from placentas using intravascular silica-bead perfusion, and then identified using shotgun proteomics. We identified 1181 plasma membrane proteins, of which 171 were enriched at the maternal blood-trophoblast interface and 192 at the fetal endothelial interface with a 70% conservation of expression in humans. Three distinct molecular subgroups of human preeclampsia were identified in existing human microarray data by using expression patterns of trophoblast-enriched proteins. Analysis of all misexpressed genes revealed divergent dysfunctions including angiogenesis (subgroup 1), MAPK signaling (subgroup 2), and hormone biosynthesis and metabolism (subgroup 3). Subgroup 2 lacked expected changes in known preeclampsia markers (sFLT1, sENG) and uniquely overexpressed GNA12. In an independent set of 40 banked placental specimens, GNA12 was overexpressed during preeclampsia when co-incident with chronic hypertension. In the current study we used a novel translational analysis to integrate mouse and human trophoblast protein expression with human microarray data. This strategy identified distinct molecular pathologies in human preeclampsia. We conclude that clinically similar preeclampsia patients exhibit divergent placental gene expression profiles thus implicating divergent molecular mechanisms in the origins of this disease.


Assuntos
Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Proteínas de Membrana/metabolismo , Placenta/metabolismo , Pré-Eclâmpsia/metabolismo , RNA Mensageiro/metabolismo , Algoritmos , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Inteligência Artificial , Teorema de Bayes , Biomarcadores/metabolismo , Endoglina , Endotélio/metabolismo , Feminino , Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/genética , Expressão Gênica , Células Gigantes/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Placenta/patologia , Pré-Eclâmpsia/diagnóstico , Pré-Eclâmpsia/genética , Gravidez , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Software , Pesquisa Translacional Biomédica , Trofoblastos/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
6.
Biol Reprod ; 86(4): 115, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22238282

RESUMO

Connexin expression and gap junctional intercellular communication (GJIC) mediated by connexin 43 (Cx43)/gap junction A1 (GJA1) are required for cytotrophoblast fusion into the syncytium, the outer functional layer of the human placenta. Cx43 also impacts intracellular signaling through protein-protein interactions. The transcription factor GCM1 and its downstream target ERVW-1/SYNCYTIN-1 are key players in trophoblast fusion and exert their actions through the ERVW-1 receptor SLC1A5/ASCT-2/RDR/ATB(0). To investigate the molecular role of the Cx43 protein and its interaction with this fusogenic pathway, we utilized stable Cx43-transfected cell lines established from the choriocarcinoma cell line Jeg3: wild-type Jeg3, alphahCG/Cx43 (constitutive Cx43 expression), JpUHD/Cx43 (doxycyclin-inducible Cx43 expression), or JpUHD/trCx43 (doxycyclin-inducible Cx43 carboxyterminal deleted). We hypothesized that truncation of Cx43 at its C-terminus would inhibit trophoblast fusion and protein interaction with either ERVW-1 or SLC1A5. In the alphahCG/Cx43 and JpUHD/Cx43 lines, stimulation with cAMP caused 1) increase in GJA1 mRNA levels, 2) increase in percentage of fused cells, and 3) downregulation of SLC1A5 expression. Cell fusion was inhibited by GJIC blockade using carbenoxylone. Neither Jeg3, which express low levels of Cx43, nor the JpUHD/trCx43 cell line demonstrated cell fusion or downregulation of SLC1A5. However, GCM1 and ERVW-1 mRNAs were upregulated by cAMP treatment in both Jeg3 and all Cx43 cell lines. Silencing of GCM1 prevented the induction of GJA1 mRNA by forskolin in BeWo choriocarcinoma cells, demonstrating that GCM1 is upstream of Cx43. All cell lines and first-trimester villous explants also demonstrated coimmunoprecipitation of SLC1A5 and phosphorylated Cx43. Importantly, SLC1A5 and Cx43 gap junction plaques colocalized in situ to areas of fusing cytotrophoblast, as demonstrated by the loss of E-cadherin staining in the plasma membrane in first-trimester placenta. We conclude that Cx43-mediated GJIC and SLC1A5 interaction play important functional roles in trophoblast cell fusion.


Assuntos
Sistema ASC de Transporte de Aminoácidos/metabolismo , Conexina 43/fisiologia , AMP Cíclico/metabolismo , Produtos do Gene env/metabolismo , Proteínas Nucleares/genética , Placenta/metabolismo , Proteínas da Gravidez/metabolismo , Fatores de Transcrição/genética , Trofoblastos/metabolismo , Fusão Celular , Linhagem Celular Tumoral , Proteínas de Ligação a DNA , Feminino , Humanos , Antígenos de Histocompatibilidade Menor , Gravidez , Primeiro Trimestre da Gravidez , Transdução de Sinais
7.
Am J Pathol ; 178(3): 1177-89, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21356369

RESUMO

Trophoblast cell invasion into the uterus is an essential process for successful pregnancy, and shallow invasion of trophoblasts into the maternal decidua is linked to preeclampsia. We have reported that Nodal, a member of the transforming growth factor-ß superfamily, acts through activin receptor-like kinase 7 (ALK7) to inhibit trophoblast proliferation and to induce apoptosis. In this study, we examined the spatial and temporal expression patterns of Nodal and ALK7 in human placenta from normal and preeclamptic pregnancies and investigated whether Nodal regulated trophoblast migration and invasion. Nodal and ALK7 were detected in villous and extravillous trophoblast cell populations in early gestation, and their levels were strongly up-regulated in preeclamptic placenta. Overexpression of Nodal or constitutively active ALK7 decreased cell migration and invasion, whereas knockdown of Nodal and ALK7 had the opposite effects. In placental explant culture, treatment with Nodal inhibited trophoblast outgrowth, whereas Nodal small-interfering RNA strongly induced the expansion of explants and the migration of extravillous trophoblast cells. Nodal stimulated the secretion of tissue inhibitor of metalloproteinase-1 and inhibited matrix metalloproteinase (MMP)-2 and MMP-9 activity. These findings suggest that the Nodal/ALK7 pathway plays important roles in human placentation and that its abnormal signaling may contribute to the development of preeclampsia.


Assuntos
Receptores de Ativinas Tipo I/metabolismo , Movimento Celular , Proteína Nodal/metabolismo , Pré-Eclâmpsia/etiologia , Transdução de Sinais , Trofoblastos/enzimologia , Trofoblastos/patologia , Feminino , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Placenta/enzimologia , Placenta/patologia , Placentação , Pré-Eclâmpsia/enzimologia , Pré-Eclâmpsia/metabolismo , Gravidez , RNA Interferente Pequeno/metabolismo , Inibidor Tecidual de Metaloproteinase-1/metabolismo
8.
Hum Reprod ; 26(5): 1025-34, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21406447

RESUMO

BACKGROUND: Severe pre-eclampsia (sPE) causes significant maternal morbidity and intrauterine growth restriction as a result of severe placental dysfunction. Defects in the formation of both extra-villous and villous trophoblast are characteristic of this disease. The outer syncytiotrophoblast layer covering the placental villi develops syncytial knots and focal necrosis while reduced invasion of the extra-villous trophoblast results in a reduced maternal blood supply and ischemia of the placental villi. The transcription factor glial cell missing-1 (GCM1) regulates formation of both types of trophoblast. GCM1 expression is reduced in placental villi of women with sPE but the functional downstream consequences of reduced GCM1 expression are unknown. METHODS AND RESULTS: In floating first trimester villous explants we demonstrated increased mRNA (2.5-fold, n = 12) and protein level (9.8-fold) of tissue inhibitor of metalloproteinase-4 (TIMP4) following repression of GCM1 (70 ± 7%) by small interfering-RNA, using RT-PCR and western blot, respectively. Similar increases in TIMP4 mRNA (4.2-fold, n = 7, P< 0.001 versus control) and protein levels were found following gene silencing of GCM1 in BeWo cells (<90% knock down of protein). TIMP4 protein was increased in placenta from women with sPE (3.5 ± 0.4 pg/µg, n = 8), compared with preterm (1.7 ± 0.17 pg/µg, n = 9) and term controls (1.6 ± 0.16 pg/µg, n = 9; P< 0.01; quantified by enzyme-linked immunosorbent assay and visualized using immunohistochemistry) with reduced GCM1 expression, mostly in the pathologic syncytial knots. CONCLUSIONS: TIMP4 is a downstream target of GCM1 that may link the consequences of reduced GCM-1-directed trophoblast differentiation to histologic and functional components of disordered placentation in sPE.


Assuntos
Vilosidades Coriônicas/metabolismo , Proteínas Nucleares/fisiologia , Pré-Eclâmpsia/metabolismo , Inibidores Teciduais de Metaloproteinases/metabolismo , Fatores de Transcrição/fisiologia , Western Blotting , Linhagem Celular , Proteínas de Ligação a DNA , Feminino , Regulação da Expressão Gênica , Humanos , Proteínas Nucleares/antagonistas & inibidores , Pré-Eclâmpsia/genética , Gravidez , Primeiro Trimestre da Gravidez/metabolismo , Interferência de RNA , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Inibidores Teciduais de Metaloproteinases/genética , Fatores de Transcrição/antagonistas & inibidores , Inibidor Tecidual 4 de Metaloproteinase
9.
Sci Rep ; 9(1): 2018, 2019 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-30765769

RESUMO

Increased inflammation and abnormal placentation are common features of a wide spectrum of pregnancy-related disorders such as intra uterine growth restriction, preeclampsia and preterm birth. The inflammatory response of the human placenta has been mostly investigated in relation to cytokine release, but the direct molecular consequences on trophoblast differentiation have not been investigated. This study measured the general effects of LPS on both extravillous and villous trophoblast physiology, and the involvement of the transcription factors PPARγ and NF-κB, specifically using 1st trimester explants and HTR-8/ SVneo cell line models. While both proteins are known for their roles in inflammatory pathways, PPARγ has been identified as an important molecule in trophoblast differentiation, suggesting its potential role in mediating a crosstalk between inflammation and trophoblast differentiation. Here, LPS (1 µg/ml) exposure of first trimester placental villous explants resulted in secretion of inflammatory cytokines, induction of apoptosis and reduction in trophoblast cell proliferation. Additionally, LPS significantly reduced expression of the trophoblast differentiation proteins GCM1 and ß-hCG, and increased invasion of the extravillous trophoblast. Activation of PPARγ by Rosiglitazone (10 µM) reversed the LPS-mediated effects on inflammatory cytokine release, trophoblast apoptosis and proliferation compared to controls. Lastly, markers of trophoblast differentiation and invasion reverted to control levels upon activation of PPARγ and concomitant inhibition of NF-κB (either by Rosiglitazone or NF-κB specific inhibitor), revealing a new role for NF-κB in trophoblast invasion. This study reveals a novel PPARγ - NF-κB axis that coordinates inflammatory and differentiation pathways in the human placenta. The ability to reverse trophoblast-associated inflammation with Rosiglitazone offers promise that the PPARγ - NF-κB pathway could one day provide a therapeutic target for placental dysfunction associated with both inflammation and abnormal trophoblast differentiation.


Assuntos
NF-kappa B/metabolismo , Placenta/efeitos dos fármacos , Placenta/lesões , Primeiro Trimestre da Gravidez/efeitos dos fármacos , Rosiglitazona/farmacologia , Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Feminino , Humanos , Inflamação/metabolismo , PPAR gama/metabolismo , Placenta/citologia , Placenta/metabolismo , Gravidez , Primeiro Trimestre da Gravidez/metabolismo , Trofoblastos/citologia , Trofoblastos/efeitos dos fármacos
10.
Methods Mol Biol ; 475: 363-82, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18979255

RESUMO

A healthy syncytium in the placenta is vital to a successful pregnancy. The trophoblast builds up the natural barrier between the mother and the developing fetus and is the site of gas, nutrition, and waste exchange. An inadequate formation of this tissue leads to several pathologies of pregnancy, which may result in fetal death during the second trimester or iatrogenic preterm delivery due to intrauterine growth restriction, preeclampsia, or abruption.Cytotrophoblastic cells fuse constantly with the overlying syncytiotrophoblast/syncytium to maintain the function of the trophoblast. Syncytin-1 is the only molecule known to directly induce fusion in the placental trophoblast. Many other proteins, such as gap junctions (e.g., connexin 40) and transcription factors, play a role in the molecular pathways directing the trophoblast turn over. Despite the significance of this process for successful placentation, the mechanisms regulating its activity remain poorly understood.In this chapter we present several different model systems that can be utilized to investigate the regulation of the cell fusion process in the trophoblast. We describe cell-based assays as well as tissue-related protocols. We show how fusion can be monitored in (1) BeWo cells as a trophoblast cell line model, (2) HEK239 using syncytin-1 as a fusion molecule, and (3) a floating villi explant model. Furthermore, we will present strategies to inhibit fusion in the different models. These techniques represent powerful tools to study the molecular mediators of cell fusion in the trophoblast.


Assuntos
Bioensaio/métodos , Fusão Celular/métodos , Modelos Biológicos , Trofoblastos/citologia , Biotinilação , Linhagem Celular , Membrana Celular/metabolismo , Produtos do Gene env/metabolismo , Humanos , Plasmídeos/genética , Proteínas da Gravidez/metabolismo , Transfecção
11.
Sci Rep ; 7(1): 5887, 2017 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-28725005

RESUMO

In severe early-onset preeclampsia (sPE) the placenta releases soluble angiogenesis-regulating proteins, trophoblast-derived fragments, and extracellular vesicles (EVs). Their relative importance in disease pathogenesis is not presently understood. We explanted placental villi from healthy and sPE women then separated the media into: total-conditioned, EV-depleted and EV-enriched media. Three fractions were compared for; angiogenic protein secretion by ELISA, angiogenic and inflammation gene mRNA expression and leukocyte adhesion assay. sPE placental villi secreted significantly less PlGF (70 ± 18 pg/mL) than preterm controls (338 ± 203; p = 0.03). sFlt-1:PlGF ratios in total-conditioned (115 ± 29) and EV-depleted media (136 ± 40) from sPE placental villi were significantly higher than in EV-enriched media (42 ± 12; p < 0.01) or any preterm or term media. Fluorescent-labeled EVs derived across normal gestation, but not from sPE, actively entered HUVECs. From sPE placental villi, the soluble fraction, but not EV-enriched fraction, significantly repressed angiogenesis (0.83 ± 0.05 fold, p = 0.02), induced HO-1 mRNA (15.3 ± 5.1 fold, p < 0.05) and induced leukocyte adhesion (2.2 ± 0.4 fold, p = 0.04). Soluble media (total-conditioned and EV-depleted media) from sPE placental villi induced endothelial dysfunction in HUVEC, while the corresponding EV-enriched fraction showed no such effects. Our data suggest that soluble factors including angiogenesis-regulating proteins, dominate the vascular pathology of this disease.


Assuntos
Endotélio/fisiopatologia , Vesículas Extracelulares/metabolismo , Pré-Eclâmpsia/fisiopatologia , Indutores da Angiogênese/metabolismo , Adesão Celular , Vilosidades Coriônicas/metabolismo , Vilosidades Coriônicas/ultraestrutura , Vesículas Extracelulares/ultraestrutura , Feminino , Fluorescência , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Inflamação/genética , Inflamação/patologia , Leucócitos/patologia , Células MCF-7 , Gravidez , Trofoblastos/metabolismo
12.
PLoS One ; 12(5): e0178056, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28545138

RESUMO

BACKGROUND: Small ubiquitin-like modifiers (SUMOs) conjugate to proteins post-translationally, thereby affecting target localization, activity and stability. Functional SUMO family members identified in the human placenta include SUMO-1 to SUMO-3, which are elevated in pre-eclampsia. Whether the fourth isoform, SUMO-4, plays a role in placental development and function remains unknown. OBJECTIVES: We tested the hypothesis that SUMO-4 is expressed in the human placenta and demonstrates altered SUMOylation in pre-eclamptic pregnancies. METHODS: SUMO-4 mRNA (qRT-PCR) and protein (Western blot and immunohistochemistry) were measured in Jar cells, BeWo cells, first trimester placental villous explants and placental tissues across normal gestation and in pre-eclampsia. SUMO-4 expression in response to oxidative stress (H2O2: 0, 0.1, 1 and 5mM), as well as, hypoxia-reperfusion (O2: 1%, 8% and 20%) was measured. Lastly, SUMO-4 binding (covalently vs. non-covalently) to target proteins was investigated. RESULTS: SUMO-4 mRNA and protein were unchanged across gestation. SUMO-4 was present in the villous trophoblast layer throughout gestation. SUMO-4 mRNA expression and protein levels were increased ~2.2-fold and ~1.8-fold in pre-eclamptic placentas compared to age-matched controls, respectively (p<0.01). SUMO-4 mRNA and protein expression increased in Jars, BeWos and first trimester placental explants with 5mM H2O2 treatment, as well as with exposure to hypoxia-reperfusion. SUMO-1 to SUMO-3 did not show consistent trends across models. SUMO-4 hyper-SUMOylation was predominantly covalent in nature. CONCLUSIONS: SUMO-4 is expressed in normal placental development. SUMO-4 expression was increased in pre-eclamptic placentas and in models of oxidative stress and hypoxic injury. These data suggests that SUMO-4 hyper-SUMOylation may be a potential post-translational mechanism in the stressed pre-eclamptic placenta.


Assuntos
Placenta/metabolismo , Pré-Eclâmpsia/metabolismo , Proteínas da Gravidez/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Hipóxia Celular , Células Cultivadas , Feminino , Humanos , Estresse Oxidativo , Placentação , Pré-Eclâmpsia/genética , Gravidez , Sumoilação , Trofoblastos/metabolismo
13.
Hypertension ; 69(1): 180-188, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27840330

RESUMO

Low molecular weight heparin (LMWH) has been investigated for the prevention of severe preeclampsia, although the mechanisms of action are unknown. The objective of this study was to investigate the cardiovascular effects of LMWH in pregnant women at high risk of preeclampsia. Pregnant women at high risk of preeclampsia (n=25) and low-risk pregnant controls (n=20) at 22 to 26 weeks' gestation underwent baseline cardiovascular assessments. High-risk women were then randomized to LMWH or saline placebo (30 mg IV bolus and 1 mg/kg subcutaneous dose). Cardiovascular function was assessed 1 and 3 hours post randomization. The in vitro endothelial effects of patient serum and exogenous LMWH on human umbilical venous endothelial cells were determined. High-risk women demonstrated a reduced cardiac output, high resistance hemodynamic profile with impaired radial artery flow-mediated dilation compared with controls. LMWH increased flow-mediated dilation in high-risk women 3 hours after randomization compared with baseline and increased plasma levels of placental growth factor, soluble fms-like tyrosine kinase-1, and myeloperoxidase. Serum from high-risk women impaired endothelial cell angiogenesis and increased PlGF-1 and PlGF-2 transcription compared with serum from low-risk controls. Coexposure of high-risk serum with LMWH improved the in vitro angiogenic response such that it was equivalent to that of low-risk serum and promoted placental growth factor secretion. LMWH improves maternal endothelial function in pregnant women at high risk of developing preeclampsia, possibly mediated through increased placental growth factor bioavailability.


Assuntos
Endotélio Vascular/fisiopatologia , Heparina de Baixo Peso Molecular/uso terapêutico , Pré-Eclâmpsia/tratamento farmacológico , Adulto , Anticoagulantes/uso terapêutico , Biomarcadores/sangue , Células Cultivadas , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/patologia , Feminino , Humanos , Pré-Eclâmpsia/sangue , Pré-Eclâmpsia/fisiopatologia , Gravidez , Proteínas da Gravidez/sangue , Fatores de Risco , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/sangue
14.
Taiwan J Obstet Gynecol ; 55(3): 385-9, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27343320

RESUMO

OBJECTIVE: To investigate the relationship between placental transthyretin (TTR) level and preeclampsia. MATERIALS AND METHODS: Placental tissues from uncomplicated and preeclamptic pregnancies were analyzed using immunohistochemistry and image analysis. We measured the mean optical density (OD) of immunohistochemical staining of TTR across multiple sections using Image Pro Plus 6.0. To avoid bias, we used placental tissue array, which contained preeclamptic placentas (n=8) and the control placentas (n=6) on the same slide. RESULTS: The mean TTR OD of the syncytiotrophoblast layer of placentas (95% confidence interval) from the first trimester was higher than those from the second/third trimester, and term placentas [0.149 (0.014-0.285) for the 1(st) trimester, 0.037 (0.000-0.073) for the 2(nd)/3(rd) trimester, and 0.011 (0.035-0.056) for term; p<0.01]. Although the OD of the second/third trimester placentas appeared greater than that of term placentas, this was not statistically significant. The mean TTR OD of the syncytiotrophoblast layer of the severe preeclampsia group was lower than that of controls [0.010 (0.005-0.016) vs. 0.027 (0.013-0.041), p<0.05]. CONCLUSION: The immunohistochemical expression of TTR in the syncytiotrophoblast layer of the placenta decreased significantly after 12 weeks of gestation, paralleling the changing demands of thyroid hormone uptake into the placenta. The reduced TTR expression in the syncytiotrophoblast layer of the preeclamptic placenta might impair thyroid hormone uptake and contribute to the pathophysiology of the disease.


Assuntos
Placenta/química , Pré-Eclâmpsia/metabolismo , Pré-Albumina/análise , Trimestres da Gravidez , Feminino , Idade Gestacional , Humanos , Gravidez , Análise Serial de Tecidos , Trofoblastos/química
15.
Reprod Sci ; 23(8): 1037-43, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26880769

RESUMO

INTRODUCTION: Apelin is a potent inotropic agent and causes endothelium-mediated vasodilation. Its cardiovascular profile suggests a role in the regulation of gestational hemodynamics. METHODS: We longitudinally assessed maternal serum apelin levels and hemodynamics (cardiac output and total peripheral resistance) between 20 and 34 weeks gestation in 18 women at high risk of placental dysfunction. Placental apelin staining was assessed by immunohistochemistry in placentas from uncomplicated pregnancies (n = 6), preterm deliveries (n = 6), preeclampsia (PET, n = 8), and isolated intrauterine growth restriction (IUGR, n = 8). Placental apelin gene expression was assessed by quantitative polymerase chain reaction. RESULTS: In the high-risk cohort, 4 fetuses developed isolated IUGR and 6 women developed PET. We obtained a median of 5 (range 2-9) hemodynamic and apelin measurements per woman. Apelin levels throughout gestation were best fitted by a quadratic curve. Apelin levels between 20 and 26 weeks gestation correlated with total peripheral resistance (r = .57, P = .01) and showed a trend toward an inverse correlation with stroke volume (r = -.42, P = .08). Apelin serum levels were 30% lower in pregnancies complicated by IUGR than in uncomplicated pregnancies or in women with preeclampsia (P = .009). Placental apelin gene expression was similar in IUGR, PET, preterm, and term normal placentas. Apelin staining was seen both in syncytiotrophoblast and stroma of the placental villi. In IUGR placentas, apelin staining was strongly decreased in both compartments compared to normals. Preeclamptic placentas showed an intermediate staining. CONCLUSIONS: Apelin levels mirror the cardiovascular changes seen in pregnancy. Serum and placental apelin levels are decreased in IUGR.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/sangue , Insuficiência Placentária/sangue , Adulto , Apelina , Pressão Sanguínea , Feminino , Retardo do Crescimento Fetal/sangue , Retardo do Crescimento Fetal/epidemiologia , Retardo do Crescimento Fetal/metabolismo , Idade Gestacional , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Estudos Longitudinais , Placenta/metabolismo , Insuficiência Placentária/epidemiologia , Insuficiência Placentária/metabolismo , Pré-Eclâmpsia/sangue , Pré-Eclâmpsia/epidemiologia , Pré-Eclâmpsia/metabolismo , Gravidez
16.
PPAR Res ; 2014: 637251, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24711815

RESUMO

Common pregnancy complications, such as severe preeclampsia and intrauterine growth restriction, disrupt pregnancy progression and impair maternal and fetal wellbeing. Placentas from such pregnancies exhibit lesions principally within the syncytiotrophoblast (SCT), a layer in direct contact with maternal blood. In humans and mice, glial cell missing-1 (GCM-1) promotes differentiation of underlying cytotrophoblast cells into the outer SCT layer. GCM-1 may be regulated by the transcription factor peroxisome proliferator-activated receptor-gamma (PPAR- γ ); in mice, PPAR- γ promotes labyrinthine trophoblast differentiation via Gcm-1, and, as we previously demonstrated, PPAR- γ activation ameliorates disease features in rat model of preeclampsia. Here, we aimed to characterize the baseline activity of PPAR- γ in the human choriocarcinoma BeWo cell line that mimics SCT formation in vitro and modulate PPAR- γ activity to study its effects on cell proliferation versus differentiation. We report a novel negative autoregulatory mechanism between PPAR- γ activity and expression and show that blocking PPAR- γ activity induces cell proliferation at the expense of differentiation, while these remain unaltered following treatment with the agonist rosiglitazone. Gaining a deeper understanding of the role and activity of PPAR- γ in placental physiology will offer new avenues for the development of secondary prevention and/or treatment options for placentally-mediated pregnancy complications.

17.
PLoS One ; 9(1): e87621, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24498154

RESUMO

BACKGROUND: Preeclampsia (PE) is characterized by exaggerated apoptosis of the villous trophoblast of placental villi. Since p53 is a critical regulator of apoptosis we hypothesized that excessive apoptosis in PE is mediated by abnormal expression of proteins participating in the p53 pathway and that modulation of the p53 pathway alters trophoblast apoptosis in vitro. METHODS: Fresh placental villous tissue was collected from normal pregnancies and pregnancies complicated by PE; Western blotting and real-time PCR were performed on tissue lysate for protein and mRNA expression of p53 and downstream effector proteins, p21, Bax and caspases 3 and 8. To further assess the ability of p53 to modulate apoptosis within trophoblast, BeWo cells and placental villous tissue were exposed to the p53-activator, Nutlin-3, alone or in combination with the p53-inhibitor, Pifithrin-α (PFT-α). Equally, Mdm2 was knocked-down with siRNA. RESULTS: Protein expression of p53, p21 and Bax was significantly increased in pregnancies complicated by PE. Conversely, Mdm2 protein levels were significantly depleted in PE; immunohistochemistry showed these changes to be confined to trophoblast. Reduction in the negative feedback of p53 by Mdm2, using siRNA and Nutlin-3, caused an imbalance between p53 and Mdm2 that triggered apoptosis in term villous explants. In the case of Nutlin, this was attenuated by Pifithrin-α. CONCLUSIONS: These data illustrate the potential for an imbalance in p53 and Mdm2 expression to promote excessive apoptosis in villous trophoblast. The upstream regulation of p53 and Mdm2, with regard to exaggerated apoptosis and autophagy in PE, merits further investigation.


Assuntos
Pré-Eclâmpsia/genética , Pré-Eclâmpsia/metabolismo , Trofoblastos/metabolismo , Trofoblastos/patologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Caspase 3/genética , Caspase 3/metabolismo , Caspase 8/genética , Caspase 8/metabolismo , Vilosidades Coriônicas/metabolismo , Vilosidades Coriônicas/patologia , Feminino , Humanos , Placenta/metabolismo , Placenta/patologia , Gravidez , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
18.
PLoS One ; 8(1): e51837, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23300953

RESUMO

The trophoblast transcription factor glial cell missing-1 (GCM1) regulates differentiation of placental cytotrophoblasts into the syncytiotrophoblast layer in contact with maternal blood. Reduced placental expression of GCM1 and abnormal syncytiotrophoblast structure are features of hypertensive disorder of pregnancy--preeclampsia. In-silico techniques identified the calcium-regulated transcriptional repressor--DREAM (Downstream Regulatory Element Antagonist Modulator)--as a candidate for GCM1 gene expression. Our objective was to determine if DREAM represses GCM1 regulated syncytiotrophoblast formation. EMSA and ChIP assays revealed a direct interaction between DREAM and the GCM1 promoter. siRNA-mediated DREAM silencing in cell culture and placental explant models significantly up-regulated GCM1 expression and reduced cytotrophoblast proliferation. DREAM calcium dependency was verified using ionomycin. Furthermore, the increased DREAM protein expression in preeclamptic placental villi was predominantly nuclear, coinciding with an overall increase in sumolylated DREAM and correlating inversely with GCM1 levels. In conclusion, our data reveal a calcium-regulated pathway whereby GCM1-directed villous trophoblast differentiation is repressed by DREAM. This pathway may be relevant to disease prevention via calcium-supplementation.


Assuntos
Regulação da Expressão Gênica , Proteínas Interatuantes com Canais de Kv/metabolismo , Neuropeptídeos/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Adulto , Animais , Cálcio/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Proteínas de Ligação a DNA , Feminino , Inativação Gênica , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Placenta/metabolismo , Gravidez , Mapeamento de Interação de Proteínas , RNA Interferente Pequeno/metabolismo , Trofoblastos/citologia
19.
Cell Calcium ; 49(5): 350-6, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21236488

RESUMO

The placenta sustains the developing fetus throughout gestation and its major functions include nutrition, gas and waste exchange via a variety of passive or active mechanisms. Up to 30 g of calcium (Ca(2+)) actively crosses the trophoblast layer during human pregnancy. The Ca(2+) ion not only plays an important role for skeletal development but is also an essential second messenger. This review is intended to highlight the implications of Ca(2+) signaling during reproduction and specifically placentation. Initially, a Ca(2+) wave induces fertilization of the oocyte. The intracellular Ca(2+) concentration is key for the blastocyst implantation, proper placental development and function. Current knowledge of many proteins involved in placental Ca(2+) regulation and their function in pathologic conditions is largely limited. Recent studies, however, point to alterations in Ca(2+) homeostasis in placental pathologies such as pre-eclampsia (PE) and intrauterine growth restriction (IUGR). A broader understanding of the role of Ca(2+) signaling during human reproduction may offer insight into impaired pregnancy outcomes.


Assuntos
Sinalização do Cálcio , Placenta/metabolismo , Blastocisto/metabolismo , Cálcio/metabolismo , Desenvolvimento Embrionário , Feminino , Fertilização , Humanos , Placenta/patologia , Placentação , Gravidez
20.
Epigenetics ; 6(3): 368-79, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21131778

RESUMO

Epigenetic processes, such as DNA methylation, are known to regulate tissue specific gene expression. We explored this concept in the placenta to define whether DNA methylation is cell-type specific. Cytotrophoblasts and fibroblasts were isolated from normal midtrimester placentas. Using immunocytochemistry, we demonstrated 95% purity for cytotrophoblasts and 60-70% for fibroblasts. We compared DNA methylation profiles from cytotrophoblasts, fibroblasts and whole placental villi using bisulfite modified genomic DNA hybridized to the Illumina Methylation27 array. Euclidean cluster analysis of the DNA methylation profiles showed 2 main clusters, one containing cytotrophoblasts and placenta, the other fibroblasts. Differential methylation analysis identified 442 autosomal CpG sites that differed between cytotrophoblasts and fibroblasts, 315 between placenta and fibroblasts and 61 between placenta and cytotrophoblasts. Three candidate methylation differences were validated by targeted pyrosequencing assays. Pyrosequencing assays were developed for CpG sites less methylated in cytotrophoblasts than fibroblasts mapping to the promoter region of the beta subunit of human chorionic gonadotropin 5 (CGB5), as well as 2 CpG sites mapping to each of 2 tumor suppressor genes. Our data suggest that epigenetic regulation of gene expression is likely to be a key factor in the functional specificity of cytotrophoblasts. These data are proof of principle for cell-type specific epigenetic regulation in placenta and demonstrate that the methylation profile of placenta is mainly driven by cytotrophoblasts.


Assuntos
Metilação de DNA , Placenta/metabolismo , Fracionamento Celular , Gonadotropina Coriônica/genética , Gonadotropina Coriônica/metabolismo , Ilhas de CpG , Epigenômica , Feminino , Humanos , Imuno-Histoquímica , Gravidez , Trofoblastos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA