Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(4)2024 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-38396719

RESUMEN

Preeclampsia (PE) is characterized by maternal hypertension and placental dysfunction, often leading to fetal growth restriction (FGR). It is associated with an overexpression of the anti-angiogenic sFLT1 protein, which originates from the placenta and serves as a clinical biomarker to predict PE. To analyze the impact of sFLT1 on placental function and fetal growth, we generated transgenic mice with placenta-specific human sFLT1 (hsFLT1) overexpression. Immunohistochemical, morphometrical, and molecular analyses of the placentas on 14.5 dpc and 18.5 dpc were performed with a focus on angiogenesis, nutrient transport, and inflammation. Additionally, fetal development upon placental hsFLT1 overexpression was investigated. Dams exhibited a mild increase in serum hsFLT1 levels upon placental hsFLT1 expression and revealed growth restriction of the fetuses in a sex-specific manner. Male FGR fetuses expressed higher amounts of placental hsFLT1 mRNA compared to females. FGR placentas displayed an altered morphology, hallmarked by an increase in the spongiotrophoblast layer and changes in labyrinthine vascularization. Further, FGR placentas showed a significant reduction in placental glycogen storage and nutrient transporter expression. Moreover, signs of hypoxia and inflammation were observed in FGR placentas. The transgenic spongiotrophoblast-specific hsFLT1 mouse line demonstrates that low hsFLT1 serum levels are sufficient to induce significant alterations in fetal and placental development in a sex-specific manner.


Asunto(s)
Retardo del Crecimiento Fetal , Preeclampsia , Ratones , Animales , Embarazo , Humanos , Masculino , Femenino , Ratones Transgénicos , Retardo del Crecimiento Fetal/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Placenta/metabolismo , Preeclampsia/genética , Inflamación/genética
2.
Development ; 143(5): 787-98, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26811378

RESUMEN

Loss of TFAP2C in mouse leads to developmental defects in the extra-embryonic compartment with lethality at embryonic day (E)7.5. To investigate the requirement of TFAP2C in later placental development, deletion of TFAP2C was induced throughout extra-embryonic ectoderm at E6.5, leading to severe placental abnormalities caused by reduced trophoblast population and resulting in embryonic retardation by E8.5. Deletion of TFAP2C in TPBPA(+) progenitors at E8.5 results in growth arrest of the junctional zone. TFAP2C regulates its target genes Cdkn1a (previously p21) and Dusp6, which are involved in repression of MAPK signaling. Loss of TFAP2C reduces activation of ERK1/2 in the placenta. Downregulation of Akt1 and reduced activation of phosphorylated AKT in the mutant placenta are accompanied by impaired glycogen synthesis. Loss of TFAP2C led to upregulation of imprinted gene H19 and downregulation of Slc38a4 and Ascl2. The placental insufficiency post E16.5 causes fetal growth restriction, with 19% lighter mutant pups. Knockdown of TFAP2C in human trophoblast choriocarcinoma JAr cells inhibited MAPK and AKT signaling. Thus, we present a model where TFAP2C in trophoblasts controls proliferation by repressing Cdkn1a and activating the MAPK pathway, further supporting differentiation of glycogen cells by activating the AKT pathway.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas Gestacionales/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Transcripción AP-2/fisiología , Animales , Diferenciación Celular , Proliferación Celular , Cruzamientos Genéticos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Femenino , Eliminación de Gen , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Genotipo , Glucógeno/metabolismo , Hibridación in Situ , Integrasas/genética , Integrasas/fisiología , Masculino , Ratones , Fosforilación , Placenta/metabolismo , Embarazo , Proteínas Gestacionales/genética , Factor de Transcripción AP-2/genética , Transgenes , Trofoblastos/metabolismo
3.
J Cell Physiol ; 234(1): 740-748, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-30078185

RESUMEN

RNA-binding proteins (RBPs) have been described for cancer cell progression and differentiation, although there is still much to learn about their mechanisms. Here, using in vivo decidualization as a model, we describe the role of RBP cold shock domain containing C2 (CSDC2) in the endometrium. Csdc2 messenger RNA expression was differentially regulated depending on time and areas of decidua development, with the most variation in antimesometrium (AM) and, to a lesser degree, in the junctional zone (JZ). Immunohistochemistry of CSDC2 showed a preferentially cytoplasmic localization at AM and JZ, and nuclear localization in underneath myometrium and mesometrium (M). Cytoplasmic localization coincided with differentiated, DESMIN-marked areas, while nuclear localization coincides with proliferative zones. Uterine suppression of CSDC2 through intrauterine-injected-specific small interfering RNA (siRNA) led to abnormal decidualization in early pregnancy, with more extended antimesometrial area and with poor M development if compared with control siRNA-injected animals. These results suggest that CSDC2 could be a regulator during decidua development.


Asunto(s)
Diferenciación Celular/genética , Endometrio/crecimiento & desarrollo , Proteínas de Unión al ARN/genética , Animales , Respuesta al Choque por Frío/genética , Citoplasma/genética , Decidua/crecimiento & desarrollo , Implantación del Embrión/genética , Endometrio/metabolismo , Femenino , Humanos , Embarazo , Dominios Proteicos/genética , ARN Interferente Pequeño/genética , Ratas , Transducción de Señal
4.
J Cell Biochem ; 118(6): 1316-1329, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27859593

RESUMEN

Since it is known that placental overexpression of the human anti-angiogenic molecule sFlt-1, the main candidate in the progression of preeclampsia, lead to intrauterine growth restriction (IUGR) in mice by lentiviral transduction of mouse blastocysts, we hypothesize that sFlt-1 influence placental morphology and physiology resulting in fetal IUGR. We therefore examined the effect of sFlt-1 on placental morphology and physiology at embryonic day 18.5 with histologic and morphometric analyses, transcript analyses, immunoblotting, and methylation studies. Interestingly, placental overexpression of sFlt-1 leads to IUGR in the fetus and results in lower placental weights. Moreover, we observed altered trophoblast differentiation with reduced expression of IGF2, resulting in a smaller placenta, a smaller labyrinth, and the loss of glycogen cells in the junctional zone. Changes in IGF2 are accompanied by small changes in its DNA methylation, whereas overall DNA methylation is unaffected. In addition, the expression of placental nutrient transporters, such as the glucose diffusion channel Cx26, is decreased. In contrast, the expression of the fatty acid transporter CD36 and the cholesterol transporter ABCA1 is significantly increased. In conclusion, placental sFlt-1 overexpression resulted in a reduction in the differentiation of the spongiotrophoblast into glycogen cells. These findings of a reduced exchange area of the labyrinth and glycogen stores, as well as decreased expression of glucose transporter, could contribute to the intrauterine growth restriction phenotype. All of these factors change the intrauterine availability of nutrients. Thus, we speculate that the alterations triggered by increased anti-angiogenesis strongly affect fetal outcome and programming. J. Cell. Biochem. 118: 1316-1329, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Retardo del Crecimiento Fetal/genética , Placenta/patología , Trofoblastos/citología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Transportador 1 de Casete de Unión a ATP/genética , Transportador 1 de Casete de Unión a ATP/metabolismo , Animales , Antígenos CD36/genética , Antígenos CD36/metabolismo , Diferenciación Celular , Conexina 26 , Conexinas/genética , Conexinas/metabolismo , Metilación de ADN , Modelos Animales de Enfermedad , Epigénesis Genética , Femenino , Retardo del Crecimiento Fetal/patología , Glucógeno/metabolismo , Humanos , Factor II del Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/metabolismo , Ratones , Placenta/metabolismo , Embarazo , Trofoblastos/metabolismo
5.
Eur J Immunol ; 46(8): 2028-42, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27184067

RESUMEN

To assess the role of alveolar macrophages (AMs) during a pulmonary Aspergillus fumigatus infection AMs were depleted by intratracheal application of diphtheria toxin (DTX) to transgenic CD11c.DTR mice prior to fungal infection. Unexpectedly, all CD11c.DTR mice treated with DTX died within 4-5 days, whether being infected with A. fumigatus or not. Despite measurable impact of DTX on lung functional parameters, these constrictions could not explain the high mortality rate. Instead, DTX-treated CD11c.DTR animals developed fulminant myocarditis (FM) characterized by massive leukocyte infiltration and myocardial cell destruction, including central parts of the heart's stimulus transmission system. In fact, standard limb lead ECG recordings of diseased but not healthy mice showed a "Brugada"-like pattern with an abnormally high ST segment pointing to enhanced susceptibility for potential lethal arrhythmias. While CD11c.DTR mice are extensively used for the characterization of CD11c(+) cells, including dendritic cells, several studies have already mentioned adverse side effects following DTX treatment. Our results demonstrate that this limitation is based on severe myocarditis but not on the expected lung constrictions, and has to be taken into consideration if this animal model is used. Based on these properties, however, the CD11c.DTR mouse might serve as useful animal model for FM.


Asunto(s)
Aspergilosis/inmunología , Antígeno CD11c/genética , Células Dendríticas/inmunología , Toxina Diftérica/toxicidad , Miocarditis/inducido químicamente , Miocardio/patología , Animales , Aspergillus fumigatus , Toxina Diftérica/administración & dosificación , Modelos Animales de Enfermedad , Electrocardiografía , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos
6.
J Lipid Res ; 56(4): 821-35, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25667419

RESUMEN

Besides bulk amounts of SM, mammalian cells produce small quantities of the SM analog ceramide phosphoethanolamine (CPE). Little is known about the biological role of CPE or enzymes responsible for CPE production. Heterologous expression studies revealed that SM synthase (SMS)2 is a bifunctional enzyme producing both SM and CPE, whereas SMS-related protein (SMSr) serves as monofunctional CPE synthase. Acute disruption of SMSr catalytic activity in cultured cells causes a rise in endoplasmic reticulum (ER) ceramides, fragmentation of ER exit sites, and induction of mitochondrial apoptosis. To address the relevance of CPE biosynthesis in vivo, we analyzed the tissue-specific distribution of CPE in mice and generated mouse lines lacking SMSr and SMS2 catalytic activity. We found that CPE levels were >300-fold lower than SM in all tissues examined. Unexpectedly, combined inactivation of SMSr and SMS2 significantly reduced, but did not eliminate, tissue-specific CPE pools and had no obvious impact on mouse development or fertility. While SMSr is widely expressed and serves as the principal CPE synthase in the brain, blocking its catalytic activity did not affect ceramide levels or secretory pathway integrity in the brain or any other tissue. Our data provide a first inventory of CPE species and CPE-biosynthetic enzymes in mammals.


Asunto(s)
Biocatálisis , Esfingomielinas/biosíntesis , Transferasas (Grupos de Otros Fosfatos Sustitutos)/metabolismo , Animales , Encéfalo/citología , Encéfalo/enzimología , Encéfalo/metabolismo , Dominio Catalítico , Supervivencia Celular , Activación Enzimática , Exones/genética , Eliminación de Gen , Regulación Enzimológica de la Expresión Génica , Hígado/citología , Hígado/enzimología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Especificidad de Órganos , Fosfatidiletanolamina N-Metiltransferasa/metabolismo , Mutación Puntual , Transporte de Proteínas , Esfingomielinas/metabolismo , Transferasas (Grupos de Otros Fosfatos Sustitutos)/química , Transferasas (Grupos de Otros Fosfatos Sustitutos)/deficiencia , Transferasas (Grupos de Otros Fosfatos Sustitutos)/genética
7.
J Cell Biochem ; 116(12): 2924-37, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26018820

RESUMEN

Several gap junction connexins have been shown to be essential for appropriate placental development and function. It is known that the expression and distribution of connexins change in response to environmental oxygen levels. The placenta develops under various oxygen levels, beginning at a low oxygen tension of approximately 2% and increasing to a tension of 8% after the onset of the uteroplacental circulation. Moreover, it has been shown that during preeclampsia (PE) placentas are subjected to chronic hypoxia. Therefore, we investigated oxygen sensitivity of placental connexins 43 and 46. Using the trophoblast cell line Jar, we demonstrated that the expression of connexin43 increased during acute hypoxia but decreased during chronic hypoxia. Chronic hypoxia resulted in the translocation of connexin43 from the membrane to the cytoplasm and in a reduction in its communication properties. In contrast, the expression of connexin46 was down-regulated during chronic hypoxia and was translocated from perinuclear areas to the cell membrane. Hypoxia-inducible factor (HIF) knockdown showed that the translocation of connexin43 but not that of connexin46 was HIF-2α dependent and was mediated by phosphoinositide 3-kinase. The up-regulation of connexin43 in combination with the down-regulation of connexin46 was confirmed in placental explants cultivated under low oxygen and in placentas with early-onset PE. Taken together, in Jar cells, placental connexins 43 and 46 are regulated during periods of low oxygen in opposite manners. The oxygen sensing of connexins in the trophoblast may play a role in physiological and pathophysiological oxygen conditions and thus may contribute to PE.


Asunto(s)
Conexina 43/biosíntesis , Conexinas/biosíntesis , Oxígeno/metabolismo , Placentación , Preeclampsia/metabolismo , Hipoxia de la Célula/genética , Línea Celular , Conexina 43/metabolismo , Conexinas/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Preeclampsia/patología , Embarazo , Trofoblastos/metabolismo
8.
Biol Reprod ; 93(2): 31, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26063869

RESUMEN

Tfap2c is required for placental development and trophoblast stem cell maintenance. Deletion of Tfap2c results in early embryonic loss because of failure in placental development. We evaluated the effect of reduced Tfap2c expression on fetal outcome and placental development. Sixty percent of the heterozygous mice were lost directly after birth. Labyrinthine differentiation was impaired, as indicated by enhanced proliferation and inclusions of cobblestone-shaped cell clusters characterized by expression of Tfap2c and glycogen stores. Moreover, expression of marker genes such as Cdx2, Eomes, Gata3, and Ascl2 are decreased in the spongiotrophoblast and indicate a lowered stem cell potential. On Day 18.5 postcoitum, the labyrinth layer of Tfap2c(+/-) placentas exhibited massive hemorrhages in the maternal blood spaces; these hemorrhages might have contributed to the significantly reduced number of live-born pups. These morphological alterations were accompanied by a shift toward sinusoidal trophoblast giant cells as the cell subpopulation lining the maternal sinusoids and toward reduction in expression of the prolactin gene family member Prl2c2, a finding characteristic of the spiral arteries lining trophoblast cells. The trophoblast stem cells heterozygous for Tfap2c exhibited a reduction in the expression level of stem cell markers and in their proliferation and differentiation capacity but did not exhibit changes in marker genes of the trophoblast giant cell lineage. Taken together, these findings indicate that a reduction in the gene dosage of placental Tfap2c leads to morphological changes in the labyrinth at midgestation and in the maternal blood spaces during late pregnancy.


Asunto(s)
Placenta/patología , Factor de Transcripción AP-2/genética , Trofoblastos/fisiología , Animales , Diferenciación Celular , Linaje de la Célula , Femenino , Marcadores Genéticos/genética , Genotipo , Células Gigantes , Ratones , Ratones Noqueados , Ratones Transgénicos , Embarazo , Prolactina/genética , Células Madre
9.
Reproduction ; 150(4): R129-36, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26150552

RESUMEN

The connexin family of proteins are best known as oligomerizing to form intercellular membrane channels (gap junctions) that metabolically and ionically couple cells to allow for coordinated cellular function. Nowhere in the body is this role better illustrated than in the uterine smooth muscle during parturition, where gap junctions conduct the contraction wave throughout the tissue to deliver the baby. Parturition is followed by the onset of lactation with connexins contributing to both the dramatic reorganization of mammary gland tissue leading up to lactation and the smooth muscle contraction of the myoepithelial cells which extrudes the milk. This review summarizes what is known about the expression and roles of individual connexin family members in the uterus during labour and in the mammary glands during development and lactation. Connexin loss or malfunction in mammary glands and the uterus can have serious implications for the health of both the mother and the newborn baby.


Asunto(s)
Conexinas/fisiología , Trabajo de Parto/fisiología , Lactancia/fisiología , Animales , Femenino , Humanos , Embarazo
10.
Cell Mol Life Sci ; 71(12): 2299-311, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24448904

RESUMEN

The CCN family of proteins consists of six high homologous matricellular proteins which act predominantly by binding to heparin sulphate proteoglycan and a variety of integrins. Interestingly, CCN proteins are regulated by ovarian steroid hormones and are able to adapt to changes in oxygen concentration, which is a necessary condition for successful implantation. CCN1 is involved in processes of angiogenesis within reproductive systems, thereby potentially contributing to diseases such as endometriosis and disturbed angiogenesis in the placenta and fetus. In the ovary, CCN2 is the key factor for follicular development, ovulation and corpora luteal luteolysis, and its deletion leads to fertility defects. CCN1, CCN2 and CCN3 seem to be regulators for human trophoblast proliferation and migration, but with CCN2 acting as a counterweight. Alterations in the expression of these three proteins could contribute to the shallow invasion properties observed in preeclampsia. Little is known about the role of CCN4-6 in the reproductive organs. The ability of CCN1, CCN2 and CCN3 to interact with numerous receptors enables them to adapt their biological function rapidly to the continuous remodelling of the reproductive organs and in the development of the placenta. The CCN proteins mediate their specific cell physiological function through the receptor type of their binding partner followed by a defined signalling cascade. Because of their partly overlapping expression patterns, they could act in a concert synergistically or in an opposite way within the reproductive organs. Imbalances in their expression levels are correlated to different human reproductive diseases, such as endometriosis and preeclampsia.


Asunto(s)
Proteínas CCN de Señalización Intercelular/fisiología , Reproducción/genética , Animales , Implantación del Embrión/genética , Endometriosis/genética , Endometriosis/metabolismo , Femenino , Humanos , Ovario/fisiología , Preeclampsia/genética , Preeclampsia/metabolismo , Embarazo , Factores Sexuales , Enfermedades Uterinas/genética , Enfermedades Uterinas/metabolismo
11.
Reproduction ; 148(1): 87-98, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24700326

RESUMEN

Oocyte quality is a well-established determinant of embryonic fate. However, the molecular participants and biological markers that affect and may predict adequate embryonic development are largely elusive. Our aim was to identify the components of the oocyte molecular machinery that part take in the production of a healthy embryo. For this purpose, we used an animal model, generated by us previously, the oocytes of which do not express Cx43 (Cx43(del/del)). In these mice, oogenesis appears normal, fertilisation does occur, early embryonic development is successful but implantation fails. We used magnetic resonance imaging analysis combined with histological examination to characterise the embryonic developmental incompetence. Reciprocal embryo transfer confirmed that the blastocyst evolved from the Cx43(del/del) oocyte is responsible for the implantation disorder. In order to unveil the genes, the impaired expression of which brings about the development of defective embryos, we carried out a genomic screening of both the oocytes and the resulting blastocysts. This microarray analysis revealed a low expression of Egr1, Rpl21 and Eif4a1 in Cx43(del/del) oocytes and downregulation of Rpl15 and Eif4g2 in the resulting blastocysts. We propose that global deficiencies in genes related to the expression of ribosomal proteins and translation initiation factors in apparently normal oocytes bring about accumulation of defects, which significantly compromise their developmental capacity. The blastocysts resulting from such oocytes, which grow within a confined space until implantation, may be unable to generate enough biological mass to allow their expansion. This information could be implicated to diagnosis and treatment of infertility, particularly to IVF.


Asunto(s)
Blastocisto/metabolismo , Implantación Tardía del Embrión/genética , Regulación del Desarrollo de la Expresión Génica , Oocitos/metabolismo , Biosíntesis de Proteínas/genética , Animales , Conexina 43/deficiencia , Conexina 43/genética , Transferencia de Embrión , Factores Eucarióticos de Iniciación/deficiencia , Factores Eucarióticos de Iniciación/genética , Femenino , Genotipo , Imagen por Resonancia Magnética , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Embarazo , Proteínas Ribosómicas/deficiencia , Proteínas Ribosómicas/genética
12.
J Magn Reson Imaging ; 39(3): 683-90, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24123570

RESUMEN

PURPOSE: To investigate the effect of daily exposure in utero to static magnetic fields during prenatal development on germ cell development and fertility of exposed offspring in adulthood. MATERIALS AND METHODS: Mice were exposed daily in utero to different static magnetic field strengths at the bore entrance or in the isocenter of 1.5 T and 7 T MRI systems during the entire course of prenatal development. RESULTS: In utero-exposed male mice revealed no effect of magnetic field strength on weight of testes and epididymis or on sperm count, sperm morphology, or fertility. Exposed pregnant female mice showed no reduced fertility in terms of pregnancy rates and litter size, pointing to a normal ovarian function. However, a reduced placental weight of offspring of intrauterine exposed female mice was observed that correlated with a decrease in embryonic weight in those animals exposed at the strongest magnetic field. This effect seemed to be parent-dependent, since it was not observed in those embryos fathered by in utero-exposed male mice. CONCLUSION: Repetitive in utero exposure to strong static magnetic fields does not impair fertility but may have a parental-dependent effect on fetal programming with regard to placental development and fetal growth.


Asunto(s)
Campos Magnéticos/efectos adversos , Placentación , Preñez , Efectos Tardíos de la Exposición Prenatal , Animales , Animales Recién Nacidos , Femenino , Infertilidad Femenina , Infertilidad Masculina , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Tamaño de los Órganos , Embarazo , Distribución Aleatoria , Espermatogénesis/fisiología , Testículo/embriología , Testículo/crecimiento & desarrollo
13.
J Magn Reson Imaging ; 39(3): 691-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24123601

RESUMEN

PURPOSE: To evaluate possible risks of strong static magnetic fields for embryo implantation, gestation, organogenesis, and embryonic development. MATERIALS AND METHODS: Pregnant mice were exposed for 75 minutes daily during the entire course of pregnancy at the bore entrance, representing the position of medical staff, and at the isocenter, representing the position of patients, of a 1.5 T and a 7 T human MRI scanner. RESULTS: No effect of static magnetic field strength was observed with regard to pregnancy rate, duration of pregnancy, litter size, still births, malformations, sex distribution, or postpartum death of offspring. During the first 8 weeks postnatal, mice exposed in utero to a magnetic field strength of 1.5 T or stronger showed a slight delay in weight gain and in time to eye opening compared to controls. CONCLUSION: Daily exposure to strong magnetic fields during pregnancy had no deleterious effect on offspring; however, a developmental retardation could be observed postnatally with regard to weight gain and eye opening.


Asunto(s)
Animales Recién Nacidos/crecimiento & desarrollo , Desarrollo Embrionario/fisiología , Campos Magnéticos/efectos adversos , Preñez , Animales , Peso al Nacer , Discapacidades del Desarrollo/etiología , Femenino , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Embarazo , Distribución Aleatoria
14.
J Cell Sci ; 124(Pt 16): 2806-15, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21807945

RESUMEN

In order to study the specific function of connexin-26 (Cx26, also known as gap junction beta-2 protein; Gjb2), we generated knockin mice that expressed either a floxed lacZ reporter or, after Cre-mediated deletion, connexin-32 (Cx32)-coding DNA, both driven by the endogenous Cx26 promoter. Heterozygous Cx26knock-inCx32 (Cx26KICx32) embryos developed normally until embryonic day 14.5 but died before birth with severe lymphedemas. Although the jugular lymph sacs were normally developed, these embryos had a strongly reduced dermal lymphatic capillary network. By analyses of ß-galactosidase reporter protein expression and lymphatic or blood endothelial-specific marker proteins, we demonstrated that Cx26 expression is temporally closely linked to lymphangiogenesis. No obvious phenotypic abnormalities were observed in Cx26KICx32 mice when Cre-mediated recombination was directed to mesenchyme or blood endothelium using the Prx1-Cre or Tie2-Cre mouse strains, respectively. By contrast, keratin-5-Cre-mediated replacement of Cx26 with Cx32 or deletion of both Cx26 alleles revealed severe lymphedemas similar to the general Cx26KICx32 phenotype. Thus, conditional ablation of Cx26 (loss of function) in ectoderm leads to partial disruption of lymphatic capillaries and embryonic death. We conclude that appropriate development of dermal lymphatic vessels in mice is dependent on the expression of Cx26 in the ectoderm.


Asunto(s)
Conexinas/metabolismo , Ectodermo/metabolismo , Endotelio Vascular/metabolismo , Linfangiogénesis , Vasos Linfáticos/embriología , Animales , Conexina 26 , Conexinas/genética , Endotelio Vascular/patología , Técnicas de Sustitución del Gen , Ingeniería Genética , Proteínas de Homeodominio/genética , Linfangiogénesis/genética , Vasos Linfáticos/patología , Linfedema/genética , Ratones , Ratones Transgénicos , Especificidad de Órganos , Regiones Promotoras Genéticas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Receptor TIE-2
15.
Biol Reprod ; 89(5): 111, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24048574

RESUMEN

Connexin43 (CX43), encoded by Gja1 in the mouse, is highly expressed in decidual cells and is known to be important for the transformation of stromal cells into the compact decidua and for neoangiogenesis. Here we investigated if the dominant Gja1(Jrt) mutation encoding CX43(G60S) in mice, which results in a phenotype resembling oculodentodigital dysplasia in humans, has an impact on decidualization, angiogenesis, and implantation. We found a reduced mean weight of fetuses at Gestational Day 17.5 in dams carrying this mutation, with the growth deficiency being independent of fetal genotype. Although the mutant implantation sites exhibited a reduction in CX43 protein, with most immunoreactivity being cytoplasmic, the decidua was morphologically intact at Embryonic Days 5.5 to 7.5. However, the mutation resulted in enhanced and irregular angiogenesis and an increased level of expression of the angiogenic factor-encoding genes Vegfa, Flt1, Kdr, and Fgf2 as well as the prolactin-related gene Prl6a. Moreover, immunolocalization of VEGFA, FLT1, and KDR revealed a homogeneous distribution pattern in the mesometrial as well as antimesometrial decidua of the mutants. Most obviously, uterine NK cells are drastically diminished in the mesometrial decidua of the mutant mice. Invasion of ectoplacental cone cells was disoriented, and placentation was established more laterally in the implantation chambers. It was concluded that the CX43(G60S) mutant impairs control of decidual angiogenesis, leading to dysmorphic placentation and fetal growth restriction. This phenomenon could contribute to the reduced fetal weights and viability of pups born of Gja1(Jrt)/+ dams.


Asunto(s)
Conexina 43/genética , Decidua/irrigación sanguínea , Neovascularización Fisiológica/genética , Placenta/citología , Placenta/fisiología , Animales , Animales Recién Nacidos , Polaridad Celular/genética , Codón sin Sentido , Femenino , Retardo del Crecimiento Fetal/genética , Genes Dominantes , Heterocigoto , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Circulación Placentaria/genética , Placentación/genética , Embarazo
16.
Mol Hum Reprod ; 19(4): 237-49, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23220688

RESUMEN

Previous studies showed that CCN3 is deregulated in early-onset pre-eclampsia (PE), a pregnancy disease associated with impaired trophoblast invasion, which leads to reduced fetal oxygen and nutrition support. Recently, we identified the glycosylated (g-CCN3) and the non-glycosylated (ng-CCN3) form of matricellular CCN3 as key factors in regulation of trophoblast proliferation and invasion. While Jeg3 cells revealed a decreased proliferation upon stimulation with both forms of CCN3, enhanced migration and invasion properties were only found for ng-CCN3. Here, we focused on the signalling cascades mitogen-activated protein kinase (MAPK), PI3 kinase/Akt and Notch/p21 for mediating the dual function of CCN3 on trophoblast proliferation versus migration in Jeg3 cells upon stimulation with g- and ng-recombinant CCN3 (g/ng-rCCN3). Analysis of the CCN3-mediated signalling pathways showed that ng-rCCN3 stimulated migration properties by activating the Akt as well as the MAPK pathway. Moreover, cell migration stimulated by ng-rCCN3 was mediated via Akt and integrin α5ß1 but not the antiproliferative effect of CCN3. There was evidence that the Notch pathway might contribute to the antiproliferative properties of both forms of CCN3 by an increase in Notch1 expression and its target gene, the cell cycle inhibitor p21. Our data showed that the presence of both forms of CCN3 is accompanied by a balance of trophoblast proliferation and migration/invasion properties, which are triggered by different signalling pathways. Thus, a deregulated expression of g/ng-CCN3 could lead to an imbalance in proliferation versus invasion, and might contribute to the shallow trophoblast invasion observed in PE.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Proteína Hiperexpresada del Nefroblastoma/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Trofoblastos/citología , Movimiento Celular , Proliferación Celular , Femenino , Humanos , Modelos Biológicos , Proteína Hiperexpresada del Nefroblastoma/metabolismo , Trofoblastos/metabolismo
17.
Biol Reprod ; 86(4): 115, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22238282

RESUMEN

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.


Asunto(s)
Sistema de Transporte de Aminoácidos ASC/metabolismo , Conexina 43/fisiología , AMP Cíclico/metabolismo , Productos del Gen env/metabolismo , Proteínas Nucleares/genética , Placenta/metabolismo , Proteínas Gestacionales/metabolismo , Factores de Transcripción/genética , Trofoblastos/metabolismo , Fusión Celular , Línea Celular Tumoral , Proteínas de Unión al ADN , Femenino , Humanos , Antígenos de Histocompatibilidad Menor , Embarazo , Primer Trimestre del Embarazo , Transducción de Señal
18.
Biol Reprod ; 87(2): 37, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22623621

RESUMEN

The nonphysiological placental oxidative environment has been implicated in many complications during human pregnancy. Oxygen tension can influence a broad spectrum of molecular changes leading to alterations in trophoblast cell lineage development. In this study, we report that mouse wild-type trophoblast stem cells (TSCs) react to low oxygen (3%) with an enhanced differentiation into the giant cell pathway, indicated by a downregulation of the early stem cell markers Eomes and Cdx2 as well as by a significant upregulation of Tfap2c and the differentiation markers Tpbpa and Prl3d1. Here we demonstrated that connexin 31/GJB3-deficient TSCs failed to stabilize HIF-1A under low oxygen, resulting in nonresponsiveness of different marker genes, such as Cdx2 and Eomes and Tfap2c and Tpbpa. Moreover, connexin 31-deficient TSCs revealed a shift in giant cell differentiation from Prl3d1 expressing parietal giant cells to Ctsq, Prl3b1, and Prl2c2-positive giant cells, probably sinusoidal and canal lining trophoblast giant cells. Thus, loss of connexin 31 led to different giant cell subtypes which bypass the progenitor regulators Tfap2c and Tpbpa under low oxygen conditions.


Asunto(s)
Diferenciación Celular , Conexinas/deficiencia , Células Madre Embrionarias/fisiología , Células Gigantes/citología , Oxígeno/metabolismo , Trofoblastos/fisiología , Animales , Linaje de la Célula , Proliferación Celular , Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción AP-2/metabolismo
19.
Biol Reprod ; 84(5): 1063-71, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21248290

RESUMEN

Though the decidua serves a critical function in implantation, the hormonal regulated pathway in decidualization is still elusive. Here we describe in detail the regional distribution and the effects of progesterone receptors (PGR), estrogen receptors (ESR), and MAPK activation on decidualization. We showed an increase in PGR A, PGR B, ESR1, and phosphorylated MAPK3-1 proteins (p-MAPK3-1), but not in ESR2, in the decidual tissue up to Day 8 of pregnancy. PGR was predominantly found in the nuclei of mesometrial decidual cells and of undifferentiated stromal cells where it colocalizes with ESR2 and ESR1. In the antimesometrial decidua, all the receptors showed cytoplasmic localization. MAPK was activated exclusively in undifferentiated stromal cells of the junctional zone between the antimesometrial and mesometrial decidua and at the border of the antimesometrial decidua. Treatment with the progesterone antagonist onapristone and/or the estrogen antagonist faslodex reduced the extent of decidual tissue and downregulated the levels of PGR and ESR1. The expression level of ESR2 was affected only by the progesterone receptor antagonist, while neither the antiprogestin nor the antiestrogen significantly modified the p-MAPK3-1 level. The inhibition of MAPK3-1 phosphorylation by PD98059 impaired the extent of decidualization and the closure reaction of the implantation chamber, and significantly downregulated ESR1. These results confirm a role of both steroid receptors in the growth and differentiation of the different decidual regions and suggest a new function for p-MAPK3-1 in regulating expression levels of ESR1, thereby maintaining the proliferation capacity of stromal cells and limiting the differentiation process in specified regions of decidual tissues.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Endometrio/citología , Endometrio/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Implantación del Embrión/efectos de los fármacos , Endometrio/efectos de los fármacos , Femenino , Antagonistas de Hormonas/farmacología , Fosforilación/efectos de los fármacos , Embarazo , Proteínas Gestacionales/antagonistas & inhibidores , Proteínas Gestacionales/metabolismo , Isoformas de Proteínas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Ratas , Ratas Wistar , Receptores de Estrógenos/antagonistas & inhibidores , Receptores de Progesterona/antagonistas & inhibidores , Regulación hacia Arriba/efectos de los fármacos
20.
Hum Reprod ; 26(10): 2850-60, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21784733

RESUMEN

BACKGROUND: Recently we have shown that the matricellular CCN3 protein expressed in invasive extravillous trophoblast cells (EVTs) is decreased in early-onset pre-eclampsia and is regulated by oxygen tension. Pathogenesis of pre-eclampsia relies on a shallow invasion of EVTs into the spiral arteries, which leads to hypoxia accompanied by uteroplacental insufficiency. Here we investigated the function of glycosylated and non-glycosylated CCN3 protein on cell growth as well as migration and invasion properties of the malignant trophoblast cell line Jeg3 which is a widely used model for the invasive trophoblast. METHODS AND RESULTS: Stable transfection of Jeg3 choriocarcinoma cells with full length CCN3 resulted in high expression of secreted glycosylated and cellular non-glycosylated CCN3. These cells revealed significantly reduced growth in cell numbers combined with a significantly increased migratory and invasive capacity. Matrix metalloprotease (MMP)-2 and MMP-9 activities were enhanced dependent on CCN3 expression, which could be confirmed by CCN3 knockdown studies. Using recombinant glycosylated and non-glycosylated CCN3, we revealed that CCN3 decreased growth in Jeg3 cell numbers independent of its glycosylation status, whereas only non-glycosylated CCN3 was able to enhance migration and invasion properties. CONCLUSIONS: The present results suggest that CCN3 protein regulates the decrease in Jeg3 cell numbers independent of its glycosylation status, whereas migratory and invasive properties are influenced only by non-glycosylated CCN3. An impaired balance in the expression of glycosylated and non-glycosylated CCN3 could contribute to the shallow invasion of EVTs observed in pre-eclampsia.


Asunto(s)
Coriocarcinoma/metabolismo , Proteína Hiperexpresada del Nefroblastoma/química , Arterias/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Glicosilación , Humanos , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/biosíntesis , Invasividad Neoplásica , Oxígeno/química , Preeclampsia/metabolismo , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA