Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Front Cell Infect Microbiol ; 13: 1138232, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37260709

RESUMEN

Human papillomaviruses (HPVs) are highly prevalent commensal viruses that require epithelial stratification to complete their replicative cycle. While HPV infections are most often asymptomatic, certain HPV types can cause lesions, that are usually benign. In rare cases, these infections may progress to non-replicative viral cycles associated with high HPV oncogene expression promoting cell transformation, and eventually cancer when not cleared by host responses. While the consequences of HPV-induced transformation on keratinocytes have been extensively explored, the impact of viral replication on epithelial homeostasis remains largely unexplored. Gap junction intercellular communication (GJIC) is critical for stratified epithelium integrity and function. This process is ensured by a family of proteins named connexins (Cxs), including 8 isoforms that are expressed in stratified squamous epithelia. GJIC was reported to be impaired in HPV-transformed cells, which was attributed to the decreased expression of the Cx43 isoform. However, it remains unknown whether and how HPV replication might impact on the expression of Cx isoforms and GJIC in stratified squamous epithelia. To address this question, we have used 3D-epithelial cell cultures (3D-EpCs), the only model supporting the productive HPV life cycle. We report a transcriptional downregulation of most epithelial Cx isoforms except Cx45 in HPV-replicating epithelia. At the protein level, HPV replication results in a reduction of Cx43 expression while that of Cx45 increases and displays a topological shift toward the cell membrane. To quantify GJIC, we pioneered quantitative gap-fluorescence loss in photobleaching (FLIP) assay in 3D-EpCs, which allowed us to show that the reprogramming of Cx landscape in response to HPV replication translates into accelerated GJIC in living epithelia. Supporting the pathophysiological relevance of our observations, the HPV-associated Cx43 and Cx45 expression pattern was confirmed in human cervical biopsies harboring HPV. In conclusion, the reprogramming of Cx expression and distribution in HPV-replicating epithelia fosters accelerated GJIC, which may participate in epithelial homeostasis and host immunosurveillance.


Asunto(s)
Carcinoma de Células Escamosas , Infecciones por Papillomavirus , Humanos , Conexinas/genética , Conexinas/metabolismo , Conexina 43/genética , Conexina 43/metabolismo , Virus del Papiloma Humano , Uniones Comunicantes/metabolismo , Epitelio , Comunicación Celular/fisiología , Transformación Celular Neoplásica
2.
Int J Mol Sci ; 24(8)2023 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-37108840

RESUMEN

It is now well established that maternal serum markers are often abnormal in fetal trisomy 21. Their determination is recommended for prenatal screening and pregnancy follow-up. However, mechanisms leading to abnormal maternal serum levels of such markers are still debated. Our objective was to help clinicians and scientists unravel the pathophysiology of these markers via a review of the main studies published in this field, both in vivo and in vitro, focusing on the six most widely used markers (hCG, its free subunit hCGß, PAPP-A, AFP, uE3, and inhibin A) as well as cell-free feto-placental DNA. Analysis of the literature shows that mechanisms underlying each marker's regulation are multiple and not necessarily directly linked with the supernumerary chromosome 21. The crucial involvement of the placenta is also highlighted, which could be defective in one or several of its functions (turnover and apoptosis, endocrine production, and feto-maternal exchanges and transfer). These defects were neither constant nor specific for trisomy 21, and might be more or less pronounced, reflecting a high variability in placental immaturity and alteration. This explains why maternal serum markers can lack both specificity and sensitivity, and are thus restricted to screening.


Asunto(s)
Síndrome de Down , Embarazo , Femenino , Humanos , Síndrome de Down/diagnóstico , Placenta/química , Gonadotropina Coriónica Humana de Subunidad beta , Biomarcadores , Diagnóstico Prenatal , Proteína Plasmática A Asociada al Embarazo , Trisomía
3.
J Am Heart Assoc ; 11(18): e026292, 2022 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-36073642

RESUMEN

Background Human cardiac biopsies are widely used in clinical and fundamental research to decipher molecular events that characterize cardiac physiological and pathophysiological states. One of the main approaches relies on the analysis of semiquantitative immunoblots that reveals alterations in protein expression levels occurring in diseased hearts. To maintain semiquantitative results, expression level of target proteins must be standardized. The expression of HKP (housekeeping proteins) is commonly used to this purpose. Methods and Results We evaluated the stability of HKP expression (actin, ß-tubulin, GAPDH, vinculin, and calsequestrin) and total protein staining within control (coefficient of variation) and comparatively with ischemic human heart biopsies (P value). All HKP exhibited a high level of intragroup (ie, actin, ß-tubulin, and GAPDH) and/or intergroup variability (ie, GAPDH, vinculin, and calsequestrin). Among all, we found total protein staining to exhibit the highest degree of stability within and between groups, which makes this reference the best to study protein expression level in human biopsies from ischemic hearts and age-matched controls. In addition, we illustrated that using an inappropriate reference protein marker misleads interpretation on SERCA2 (sarco/endoplasmic reticulum Ca2+ ATPase) and cMyBPC (cardiac myosin binding protein-C) expression level after myocardial infarction. Conclusions These reemphasize the need to standardize the level of protein expression with total protein staining in comparative immunoblot studies on human samples from control and diseased hearts.


Asunto(s)
Actinas , Calsecuestrina , Miosinas Cardíacas , Isquemia , Actinas/metabolismo , Biopsia , Miosinas Cardíacas/metabolismo , Grupos Control , Humanos , Tubulina (Proteína)/metabolismo , Vinculina/metabolismo
4.
Cells ; 10(4)2021 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-33923648

RESUMEN

Under physiological conditions, cAMP signaling plays a key role in the regulation of cardiac function. Activation of this intracellular signaling pathway mirrors cardiomyocyte adaptation to various extracellular stimuli. Extracellular ligand binding to seven-transmembrane receptors (also known as GPCRs) with G proteins and adenylyl cyclases (ACs) modulate the intracellular cAMP content. Subsequently, this second messenger triggers activation of specific intracellular downstream effectors that ensure a proper cellular response. Therefore, it is essential for the cell to keep the cAMP signaling highly regulated in space and time. The temporal regulation depends on the activity of ACs and phosphodiesterases. By scaffolding key components of the cAMP signaling machinery, A-kinase anchoring proteins (AKAPs) coordinate both the spatial and temporal regulation. Myocardial infarction is one of the major causes of death in industrialized countries and is characterized by a prolonged cardiac ischemia. This leads to irreversible cardiomyocyte death and impairs cardiac function. Regardless of its causes, a chronic activation of cardiac cAMP signaling is established to compensate this loss. While this adaptation is primarily beneficial for contractile function, it turns out, in the long run, to be deleterious. This review compiles current knowledge about cardiac cAMP compartmentalization under physiological conditions and post-myocardial infarction when it appears to be profoundly impaired.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Miocardio/patología , Transducción de Señal , Animales , Humanos , Receptores Acoplados a Proteínas G/metabolismo
6.
Endocrinology ; 160(3): 684-698, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30715257

RESUMEN

Placental development is particularly altered in trisomy of chromosome 21 (T21)-affected pregnancies. We previously described in T21-affected placentae an abnormal paracrine crosstalk between the villus mesenchymal core and villus trophoblasts. T21-affected placentae are known to be characterized by their hypovascularity. However, the causes of this anomaly remain not fully elucidated. Therefore, the hypothesis of an abnormal paracrine crosstalk between fetal mesenchymal core and placental endothelial cells (PLECs) was evocated. Villus mesenchymal cells from control (CMCs) and T21 placentae (T21MCs) were isolated and grown in culture to allow their characterization and collection of conditioned media for functional analyses (CMC-CM and T21MC-CM, respectively). Interestingly, PLEC proliferation and branching ability were less stimulated by T21MC-CM than by CMC-CM. Protein array analysis identified secreted proangiogenic growth factors in CMC-CM, which were reduced in T21MC-CM. Combined mass spectrometry and biochemical analysis identified spondin-2 as a factor decreased in T21MC-CM compared with CMC-CM. We found that exogenous spondin-2 stimulated PLEC proliferation and established that T21MC-CM supplemented with spondin-2 recovered conditioned media ability to induce PLEC proliferation and angiogenesis. Hence, this study demonstrates a crosstalk between villus mesenchymal and fetal endothelial cells, in which spondin-2 secreted from mesenchymal cells plays a central role in placental vascular functions. Furthermore, our results also suggest that a reduction in spondin-2 secretion may contribute to the pathogenesis of T21 placental hypovascularity.


Asunto(s)
Síndrome de Down/fisiopatología , Proteínas de la Matriz Extracelular/fisiología , Proteínas de Neoplasias/fisiología , Placenta/irrigación sanguínea , Placentación , Estudios de Casos y Controles , Endostatinas/metabolismo , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neovascularización Fisiológica , Placenta/fisiopatología , Embarazo
7.
Biochem J ; 475(2): 455-476, 2018 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-29259079

RESUMEN

A limited number of human cells can fuse to form multinucleated syncytia. In the differentiation of human placenta, mononuclear cytotrophoblasts fuse to form an endocrinologically active, non-proliferative, multinucleated syncytium. This syncytium covers the placenta and manages the exchange of nutrients and gases between maternal and fetal circulation. We recently reported protein kinase A (PKA) to be part of a macromolecular signaling complex with ezrin and gap junction protein connexin 43 (Cx43) that provides cAMP-mediated control of gap junction communication. Here, we examined the associated phosphorylation events. Inhibition of PKA activity resulted in decreased Cx43 phosphorylation, which was associated with reduced trophoblast fusion and differentiation. In vitro studies using peptide arrays, together with mass spectrometry, pointed to serine 369 and 373 of Cx43 as the major PKA phosphorylation sites that increases gap junction assembly at the plasmalemma. A combination of knockdown and reconstitution experiments and gap-fluorescence loss in photobleaching assays with mutant Cx43 containing single or double phosphoserine-mimicking amino acid substitutions in putative PKA phosphorylation sites demonstrated that phosphorylation of S369 and S373 mediated gap junction communication, trophoblast differentiation, and cell fusion.


Asunto(s)
Conexina 43/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas del Citoesqueleto/metabolismo , Uniones Comunicantes/metabolismo , Serina/metabolismo , Trofoblastos/metabolismo , Adulto , Secuencia de Aminoácidos , Comunicación Celular/genética , Diferenciación Celular , Fusión Celular , Membrana Celular/química , Membrana Celular/metabolismo , Cesárea , Conexina 43/genética , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas del Citoesqueleto/genética , Femenino , Uniones Comunicantes/ultraestructura , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Humanos , Péptidos/síntesis química , Péptidos/metabolismo , Fosforilación , Embarazo , Cultivo Primario de Células , Transducción de Señal , Trofoblastos/citología
8.
Sci Rep ; 7: 42173, 2017 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-28176826

RESUMEN

Only a limited number of human cells can fuse to form a multinucleated syncytium. Cell fusion occurs as part of the differentiation of some cell types, including myotubes in muscle and osteoclasts in remodeling bone. In the differentiation of the human placenta, mononuclear cytotrophoblasts aggregate and fuse to form endocrinologically active, non-proliferative, multinucleated syncytia. These syncytia allow the exchange of nutrients and gases between the maternal and fetal circulation. Alteration of syncytial formation during pregnancy affects fetal growth and the outcome of the pregnancy. Here, we demonstrate the role of annexin A5 (AnxA5) in syncytial formation by cellular delivery of recombinant AnxA5 and RNA interference. By a variety of co-immunoprecipitation, immunolocalization and proximity experiments, we show that a pool of AnxA5 organizes at the inner-leaflet of the plasma membrane in the vicinity of a molecular complex that includes E-Cadherin, α-Catenin and ß-Catenin, three proteins previously shown to form adherens junctions implicated in cell fusion. A combination of knockdown and reconstitution experiments with AnxA5, with or without the ability to self-assemble in 2D-arrays, demonstrate that this AnxA5 2D-network mediates E-Cadherin mobility in the plasmalemma that triggers human trophoblasts aggregation and thereby cell fusion.


Asunto(s)
Anexina A5/genética , Cadherinas/genética , Membrana Celular/metabolismo , Células Gigantes/metabolismo , Trofoblastos/metabolismo , alfa Catenina/genética , beta Catenina/genética , Uniones Adherentes/metabolismo , Uniones Adherentes/ultraestructura , Adulto , Anexina A5/antagonistas & inhibidores , Anexina A5/metabolismo , Antígenos CD , Cadherinas/metabolismo , Comunicación Celular , Diferenciación Celular , Fusión Celular , Movimiento Celular , Femenino , Regulación de la Expresión Génica , Células Gigantes/citología , Humanos , Embarazo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Trofoblastos/citología , alfa Catenina/metabolismo , beta Catenina/metabolismo
9.
Cell Signal ; 32: 1-11, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28077322

RESUMEN

Communication between adjacent cells can occur via gap junctions (GJ) composed of connexin (Cx) hexamers that allow passage of small molecules. One of the most widely and highly expressed Cxs in human tissues is Cx43, shown to be regulated through phosphorylation by several kinases including PKA. Ezrin is a membrane associated protein that can serve as an A-kinase anchoring protein (AKAP) and hold an anchored pool of PKA. Here, we used the liver epithelial cell line IAR20, which expresses Cx43 as the predominant GJ protein, to test the hypothesis that Ezrin may associate with Cx43 in cell types that form stable GJs and serve as an AKAP. Our biochemical and proteomics data indicate that Ezrin associates with Cx43 in epithelial cells. Analyses by confocal immunofluorescence microscopy and proximity ligation assays demonstrate that Ezrin and Cx43 co-localize, together with zonula occludens-1 (ZO-1) and PKA RIα and RIIα, at the cell membrane. Quantitative gap-FRAP experiments show increased GJ intercellular communication after cAMP stimulation. Moreover, loading of cells with the Ht31 peptide that displaces both PKA RIα and RIIα from the AKAP or a peptide that disrupts the Cx43-Ezrin interaction reverts the effect and reduces the level of communication, supporting the hypothesis that in IAR20 cells Ezrin associates with Cx43 (in complex with ZO-1) which places PKA in proximity to Cx43, enabling its phosphorylation and GJ opening.


Asunto(s)
Comunicación Celular , Conexina 43/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas del Citoesqueleto/metabolismo , Células Epiteliales/metabolismo , Uniones Comunicantes/metabolismo , Hígado/citología , Animales , Membrana Celular/metabolismo , Complejos Multiproteicos/metabolismo , Fosforilación , Ratas
10.
Biol Aujourdhui ; 210(3): 139-151, 2016.
Artículo en Francés | MEDLINE | ID: mdl-27813475

RESUMEN

In the human placenta, mononuclear cytotrophoblasts fuse to form multinucleated syncytia ensuring hormonal production and nutrient exchanges between the maternal and fetal circulation. The syncytial formation is necessary for the maintenance of pregnancy and for fetal growth. The cAMP signaling pathway is the major route to trigger trophoblast fusion and its activation results in phosphorylation of specific intracellular target proteins and assembly of macromolecular protein complexes constituting the fusogenic machinery at the plasma membrane. Specificity in cAMP signaling is ensured by generation of localized pools of cAMP controlled by cAMP phosphodiesterases (PDEs) and by discrete spatial and temporal activation of protein kinase A (PKA) in supramolecular signaling clusters inside the cell organized by A-kinase-anchoring proteins (AKAPs) and by organization of signal termination by protein phosphatases (PPs). Here we summarize the current knowledge of proteins and protein macrocomplexes involved in the spatiotemporal regulation of cAMP signaling that triggers human trophoblast fusion.


Asunto(s)
AMP Cíclico/fisiología , Trofoblastos/fisiología , 3',5'-AMP Cíclico Fosfodiesterasas/fisiología , Fusión Celular , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Femenino , Humanos , Placentación/fisiología , Embarazo , Transducción de Señal/fisiología
11.
Hypertension ; 68(6): 1438-1446, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27698065

RESUMEN

The effects of fluid shear stress (FSS) on the human syncytiotrophoblast and its biological functions have never been studied. During pregnancy, the syncytiotrophoblast is the main source of placental growth factor (PlGF), a proangiogenic factor involved in the placental angiogenesis and the vascular adaptation to pregnancy. The role of FSS in regulating PlGF expression in syncytiotrophoblasts is unknown. We investigated the impact of FSS on the production and secretion of the PlGF by the human syncytiotrophoblasts in primary cell culture. Laminar and continuous FSS (1 dyn cm-2) was applied to human syncytiotrophoblasts cultured in a parallel-plate flow chambers. Secreted levels of PlGF, sFlt-1 (soluble fms-like tyrosin kinase-1), and prostaglandin E2 were tested by immunologic assay. PlGF levels of mRNA and intracellular protein were examined by RT-PCR and Western blot, respectively. Intracellular cAMP levels were examined by time-resolved fluorescence resonance energy transfer cAMP accumulation assay. Production of cAMP and PlGF secretion was significantly increased in FSS conditions compared with static conditions. Western blot analysis of cell extracts exposed to FSS showed an increased phosphorylation of protein kinase A substrates and cAMP response element-binding protein on serine 133. FSS-induced phosphorylation of cAMP response element-binding protein and upregulation of PlGF were prevented by inhibition of protein kinase A with H89 (3 µmol/L). FSS also triggers intracellular calcium flux, which increases the synthesis and release of prostaglandin E2. The enhanced intracellular cAMP in FSS conditions was blocked by COX1/COX2 (cyclooxygenase) inhibitors, suggesting that the increase in prostaglandin E2 production could activate the cAMP/protein kinase A pathway in an autocrine/paracrine fashion. FSS activates the cAMP/protein kinase A pathway leading to upregulation of PlGF in human syncytiotrophoblast.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas de la Membrana/genética , Factor de Crecimiento Placentario/genética , Análisis de Varianza , Células Cultivadas , Femenino , Humanos , Immunoblotting , Inmunohistoquímica , Placenta/metabolismo , Placenta/fisiopatología , Embarazo , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Transducción de Señal/genética , Estrés Mecánico , Trofoblastos/metabolismo , Regulación hacia Arriba
12.
Front Pharmacol ; 6: 202, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26441659

RESUMEN

During human placentation, mononuclear cytotrophoblasts fuse to form multinucleated syncytia ensuring hormonal production and nutrient exchanges between the maternal and fetal circulation. Syncytial formation is essential for the maintenance of pregnancy and for fetal growth. The cAMP signaling pathway is the major route to trigger trophoblast fusion and its activation results in phosphorylation of specific intracellular target proteins, in transcription of fusogenic genes and assembly of macromolecular protein complexes constituting the fusogenic machinery at the plasma membrane. Specificity in cAMP signaling is ensured by generation of localized pools of cAMP controlled by cAMP phosphodiesterases (PDEs) and by discrete spatial and temporal activation of protein kinase A (PKA) in supramolecular signaling clusters inside the cell organized by A-kinase-anchoring proteins (AKAPs) and by organization of signal termination by protein phosphatases (PPs). Here we present original observations on the available components of the cAMP signaling pathway in the human placenta including PKA, PDE, and PP isoforms as well as AKAPs. We continue to discuss the current knowledge of the spatiotemporal regulation of cAMP signaling triggering trophoblast fusion.

13.
Commun Integr Biol ; 8(4): e1057361, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26478781

RESUMEN

Anchored protein kinase A (PKA) bound to A Kinase Anchoring Protein (AKAP) mediates effects of localized increases in cAMP in defined subcellular microdomains and retains the specificity in cAMP-PKA signaling to distinct extracellular stimuli. Gap junctions are pores between adjacent cells constituted by connexin proteins that provide means of communication and transfer of small molecules. While the PKA signaling is known to promote human trophoblast cell fusion, the gap junction communication through connexin 43 (Cx43) is a prerequisite for this process. We recently demonstrated that trophoblast fusion is regulated by ezrin, a known AKAP, which binds to Cx43 and delivers PKA in the vicinity gap junctions. We found that disruption of the ezrin-Cx43 interaction abolished PKA-dependent phosphorylation of Cx43 as well as gap junction communication and subsequently cell fusion. We propose that the PKA-ezrin-Cx43 macromolecular complex regulating gap junction communication constitutes a general mechanism to control opening of Cx43 gap junctions by phosphorylation in response to cAMP signaling in various cell types.

14.
PLoS One ; 10(7): e0133506, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26186596

RESUMEN

The chorionic villus of the human placenta is the source of specific endocrine functions and nutrient exchanges. These activities are ensured by the syncytiotrophobast (ST), which bathes in maternal blood. The ST arises and regenerates throughout pregnancy by fusion of underlying cytotrophoblasts (CT). Any anomaly of ST formation or regeneration can affect pregnancy outcome and fetal growth. Because of its direct interaction with maternal blood, the ST is sensitive to drugs, pollutants and xenohormones. Ex vivo assays of perfused cotyledon show that formaldehyde, a common pollutant present in furniture, paint and plastics, can accumulate in the human placenta and cross to the fetal compartment. By means of RT-qPCR, immunoblot and immunocytochemistry experiments, we demonstrate in vitro that formaldehyde exerts endocrine toxicity on human trophoblasts, including a decrease in the production of protein hormones of pregnancy. In addition, formaldehyde exposure triggered human trophoblast fusion by upregulating syncitin-1 receptor expression (ASC-type amino-acid transporter 2: ASCT2). Moreover, we show that formaldehyde-exposed trophoblasts present an altered redox status associated with oxidative stress, and an increase in ASCT2 expression intended to compensate for this stress. Finally, we demonstrate that the adverse effects of formaldehyde on trophoblast differentiation and fusion are reversed by N-acetyl-L-cysteine (Nac), an antioxidant.


Asunto(s)
Diferenciación Celular , Formaldehído/toxicidad , Hormonas Placentarias/metabolismo , Trofoblastos/efectos de los fármacos , Adulto , Sistema de Transporte de Aminoácidos ASC/genética , Sistema de Transporte de Aminoácidos ASC/metabolismo , Células Cultivadas , Femenino , Humanos , Intercambio Materno-Fetal , Antígenos de Histocompatibilidad Menor , Estrés Oxidativo , Circulación Placentaria , Embarazo , Trofoblastos/citología , Trofoblastos/metabolismo
15.
Reprod Biol Endocrinol ; 12: 129, 2014 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-25543334

RESUMEN

BACKGROUND: Pregnancy-associated placental protein-A (PAPP-A) is a metalloprotease which circulates as an hetero-tetramer in maternal blood. Its maternal serum concentration in fetal trisomy 21 is decreased during the first trimester, so that PAPP-A is a useful screening biomarker. However, the regulation of PAPP-A placental secretion is unclear. We therefore investigated the secretion of PAPP-A in pregnancies complicated by fetal aneuploidies, both in vivo and in vitro. METHODS: Maternal serum collected between 10 WG and 33 WG during 7014 normal pregnancies and 96 pregnancies complicated by fetal trisomy 21, 18, and 13 were assayed for PAPP-A using the Immulite 2000xpi system®. The pregnancies were monitored using ultrasound scanning, fetal karyotyping and placental analysis. Villous cytotrophoblasts were isolated from normal and trisomic placenta and cultured to investigate PAPP-A secretion in vitro (n=6). RESULTS: An increased nuchal translucency during the first trimester is a common feature of many chromosomal defect but each aneuploidy has its own syndromic pattern of abnormalities detectable at the prenatal ultrasound scanning and confirmed at the fetal examination thereafter. PAPP-A levels rise throughout normal pregnancy whereas in trisomy 21, PAPP-A levels were significantly decreased, but only during the first trimester. PAPP-A levels were decreased in trisomy 13 and sharply in trisomy 18, whatever the gestational age. In vitro, PAPP-A secretion was decreased in aneuploidy, and associated with decreased hCG secretion in Trisomy 21 and 18. These biochemical profiles did not appear to be linked to any specific histological lesions affecting the placenta. CONCLUSIONS: These profiles may reflect different quantitative and qualitative placental dysfunctions in the context of these aneuploidies.


Asunto(s)
Enfermedades Fetales/genética , Complicaciones del Embarazo , Proteína Plasmática A Asociada al Embarazo/metabolismo , Trisomía/genética , Células Cultivadas , Gonadotropina Coriónica/sangre , Gonadotropina Coriónica/metabolismo , Trastornos de los Cromosomas/genética , Cromosomas Humanos Par 13/genética , Cromosomas Humanos Par 18/genética , Estudios de Cohortes , Síndrome de Down/genética , Femenino , Enfermedades Fetales/diagnóstico , Enfermedades Fetales/diagnóstico por imagen , Edad Gestacional , Humanos , Medida de Translucencia Nucal/métodos , Placenta/citología , Placenta/metabolismo , Embarazo , Trisomía/diagnóstico , Síndrome de la Trisomía 13 , Trofoblastos/citología , Trofoblastos/metabolismo
16.
J Cell Sci ; 127(Pt 19): 4172-85, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25052094

RESUMEN

Cell fusion occurs as part of the differentiation of some cell types, including myotubes in muscle and osteoclasts in remodeling bone. In the human placenta, mononuclear cytotrophoblasts in a human chorionic gonadotropin (hCG)-driven process fuse to form multinucleated syncytia that allow the exchange of nutrients and gases between the maternal and fetal circulation. Experiments in which protein kinase A (PKA) is displaced from A-kinase anchoring proteins (AKAPs), or in which specific AKAPs are depleted by siRNA-mediated knockdown, point to ezrin as a scaffold required for hCG-, cAMP- and PKA-mediated regulation of the fusion process. By a variety of immunoprecipitation and immunolocalization experiments, we show that ezrin directs PKA to a molecular complex of connexin 43 (Cx43, also known as GJA1) and zona occludens-1 (ZO-1, also known as TJP1). A combination of knockdown experiments and reconstitution with ezrin or Cx43 with or without the ability to bind to its interaction partner or to PKA demonstrate that ezrin-mediated coordination of the localization of PKA and Cx43 is necessary for discrete control of Cx43 phosphorylation and hCG-stimulated gap junction communication that triggers cell fusion in cytotrophoblasts.


Asunto(s)
Conexina 43/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas del Citoesqueleto/metabolismo , Uniones Comunicantes/metabolismo , Trofoblastos/metabolismo , Comunicación Celular/fisiología , Diferenciación Celular , Fusión Celular , Femenino , Humanos , Proteínas de la Membrana/metabolismo , Embarazo , Transducción de Señal , Trofoblastos/citología
17.
EMBO J ; 30(21): 4371-86, 2011 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-21983901

RESUMEN

Adrenergic stimulation of adipocytes yields a cAMP signal that activates protein kinase A (PKA). PKA phosphorylates perilipin, a protein localized on the surface of lipid droplets that serves as a gatekeeper to regulate access of lipases converting stored triglycerides to free fatty acids and glycerol in a phosphorylation-dependent manner. Here, we report a new function for optic atrophy 1 (OPA1), a protein known to regulate mitochondrial dynamics, as a dual-specificity A-kinase anchoring protein associated with lipid droplets. By a variety of protein interaction assays, immunoprecipitation and immunolocalization experiments, we show that OPA1 organizes a supramolecular complex containing both PKA and perilipin. Furthermore, by a combination of siRNA-mediated knockdown, reconstitution experiments using full-length OPA1 with or without the ability to bind PKA or truncated OPA1 fused to a lipid droplet targeting domain and cellular delivery of PKA anchoring disruptor peptides, we demonstrate that OPA1 targeting of PKA to lipid droplets is necessary for hormonal control of perilipin phosphorylation and lipolysis.


Asunto(s)
Agonistas Adrenérgicos beta/farmacología , GTP Fosfohidrolasas/fisiología , Metabolismo de los Lípidos/genética , Lipólisis/efectos de los fármacos , Células 3T3-L1 , Proteínas de Anclaje a la Quinasa A/genética , Proteínas de Anclaje a la Quinasa A/metabolismo , Proteínas de Anclaje a la Quinasa A/fisiología , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , GTP Fosfohidrolasas/antagonistas & inhibidores , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Isoproterenol/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Lipólisis/genética , Ratones , Modelos Biológicos , Perilipina-1 , Fosfoproteínas/metabolismo , ARN Interferente Pequeño/farmacología , Receptores Adrenérgicos beta/metabolismo , Receptores Adrenérgicos beta/fisiología
18.
Endocrinology ; 152(12): 5017-28, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21952245

RESUMEN

Placental development is markedly abnormal in trisomy 21 (T21) pregnancies. We hypothesized that abnormal paracrine cross talk between the fetal mesenchymal core and the trophoblast might be involved in the defect of syncytiotrophoblast formation and function. In a large series of primary cultured human cytotrophoblasts isolated from second-trimester control (n = 44) and T21 placentae (n = 71), abnormal trophoblast fusion and differentiation was observed in more than 90% of T21 cases. We then isolated and cultured villous mesenchymal cells from control (n = 10) and T21 placentae (n = 8) and confirmed their fetal origin. Conditioned medium of control mesenchymal cells overcame the abnormal trophoblast fusion of T21 cytotrophoblasts by activating the TGFß signaling pathway, as shown by the phosphospecific protein microarray analysis and the use of TGFß signaling pathway antagonists. Using protein arrays, we further analyzed the cytokines present in the conditioned medium from control and T21 mesenchymal cells. Activin-A was identified as strongly secreted by cells from both sources, but at a significantly (P < 0.01) lower level in the case of T21 mesenchymal cells. Recombinant activin-A stimulated T21 trophoblast fusion. Blocking activin-A antibody inhibited the fusion induced by conditioned medium and exogenous activin-A. Furthermore, follistatin, an activin-A binding protein largely secreted by T21 mesenchymal cells, inhibited the conditioned medium fusogenic activity. These results show that the defective trophoblast fusion and differentiation associated with T21 can be overcome in vitro and reveal the key role of the fetal mesenchymal core in human trophoblast differentiation.


Asunto(s)
Activinas/farmacología , Síndrome de Down/tratamiento farmacológico , Mesodermo/química , Comunicación Paracrina/efectos de los fármacos , Trofoblastos/patología , Diferenciación Celular , Fusión Celular , Células Cultivadas , Citocinas/análisis , Femenino , Feto , Humanos , Mesodermo/citología , Mesodermo/fisiología , Placenta , Embarazo , Factor de Crecimiento Transformador beta/metabolismo , Trofoblastos/citología , Trofoblastos/efectos de los fármacos
19.
Adv Exp Med Biol ; 714: 103-12, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21506009

RESUMEN

Trophoblastic cell fusion is one essential step of the human trophoblast differentiation leading to formation of the syncytiotrophoblast, site of the numerous placental functions. This process is multifactorial and finely regulated. Using the physiological model of primary culture of trophoblastic cells isolated from human placenta, we have identified different membrane proteins directly involved in trophoblastic cell fusion: connexin 43, ZO-1 and recently syncytins. These fusogenic membrane retroviral envelop glycoproteins: syncytin-1 (encoded by the HERV-W gene) and syncytin-2 (encoded by the FRD gene) and their receptors are major factors involved in human placental development. Disturbances of syncytiotrophoblast formation are observed in trisomy 21-affected placentas. Overexpression of the copper/zinc superoxide dismutase (SOD-1), encoded by chromosome 21 as well as an abnormal hCG signaling are implicated in the defect of syncytiotrophoblast formation. This abnormal trophoblast fusion and differentiation in trisomy 21-affected placenta is reversible in vitro by different ways.


Asunto(s)
Síndrome de Down/patología , Trofoblastos/patología , Diferenciación Celular , Fusión Celular , Productos del Gen env/fisiología , Humanos , Proteínas Gestacionales/fisiología
20.
Am J Physiol Cell Physiol ; 298(6): C1517-26, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20200207

RESUMEN

Trophoblastic cell-cell fusion is an essential event required during human placental development. Several membrane proteins have been described to be directly involved in this process, including connexin 43 (Cx43), syncytin 1 (Herv-W env), and syncytin 2 (Herv-FRD env glycoprotein). Recently, zona occludens (ZO) proteins (peripheral membrane proteins associated with tight junctions, adherens junctions, and gap junctions) were shown to be involved in mouse placental development. Moreover, zona occludens 1 (ZO-1) was localized mainly at the intercellular boundaries between human trophoblastic cells. Therefore the role of ZO-1 in the dynamic process of human trophoblastic cell-cell fusion was investigated using primary trophoblastic cells in culture. In vitro as in situ, ZO-1 was localized mainly at the intercellular boundaries between trophoblastic cells where its expression substantially decreased during differentiation and during fusion. At the same time, Cx43 was localized at the interface of trophoblastic cells and its expression increased during differentiation. To determine a functional role for ZO-1 during trophoblast differentiation, small interfering RNA (siRNA) was used to knock down ZO-1 expression. Cytotrophoblasts treated with ZO-1 siRNA fused poorly, but interestingly, decreased Cx43 expression without altering the functionality of trophoblastic cell-cell communication as measured by relative permeability time constant determined using gap-FRAP experiments. Because kinetics of Cx43 and ZO-1 proteins show a mirror image, a potential association of these two proteins was investigated. By using coimmunoprecipitation experiments, a physical interaction between ZO-1 and Cx43 was demonstrated. These results demonstrate that a decrease in ZO-1 expression reduces human trophoblast cell-cell fusion and differentiation.


Asunto(s)
Diferenciación Celular , Fusión Celular , Uniones Intercelulares/metabolismo , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Placenta/metabolismo , Trofoblastos/metabolismo , Forma de la Célula , Células Cultivadas , Gonadotropina Coriónica/metabolismo , Conexina 43/metabolismo , Desmoplaquinas/metabolismo , Femenino , Recuperación de Fluorescencia tras Fotoblanqueo , Edad Gestacional , Humanos , Inmunohistoquímica , Inmunoprecipitación , Cinética , Proteínas de la Membrana/genética , Fosfoproteínas/genética , Placenta/citología , Embarazo , Unión Proteica , Interferencia de ARN , Factores de Tiempo , Proteína de la Zonula Occludens-1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...