Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Placenta ; 36(11): 1260-5, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26390805

RESUMO

INTRODUCTION: Netrin-4 is a secreted member of the laminin-related protein family, known to be involved in axonal guidance and endothelial cell survival, proliferation, and migration. We have recently reported the cellular localization of netrin-4 and its receptor neogenin in human first trimester and term placenta. A strong expression of netrin-4 was observed in trophoblast and in endothelial cells, suggesting a potential role of this protein in placental angiogenesis. In relation to human pregnancy, it has been reported that circulating netrin-4 were increased in fetal umbilical cord blood of intrauterine growth restriction IUGR compared to normal pregnancy suggesting an adverse effect of this protein on placental and fetal development. The aim of this study was to determine the role of netrin-4 in placental angiogenesis. METHODS: The effects of netrin-4 on proliferation, migration, tube-like organization, and spheroid sprouting of human placental microvascular endothelial cells (HPEC) were studied. RESULTS: We demonstrated that netrin-4 inhibits HPEC proliferation, tube-like formation, migration and spheroid sprouting, suggesting a direct role of netrin-4 in the regulation of intra-villus angiogenesis. DISCUSSION: This is the first report of an anti-angiogenic activity of netrin-4 in human placenta. This study brings new insights into netrin-4 roles in placental angiogenesis and suggests possible involvements of netrin-4 in angiogenesis-related pathologies such as IUGR.


Assuntos
Células Endoteliais/fisiologia , Neovascularização Fisiológica , Fatores de Crescimento Neural/fisiologia , Movimento Celular , Proliferação de Células , Células Cultivadas , Humanos , Netrinas , Esferoides Celulares/fisiologia
2.
Biol Reprod ; 91(3): 73, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25122063

RESUMO

EG-VEGF is an angiogenic factor that we identified as a new placental growth factor during human pregnancy. EG-VEGF is also expressed in the mouse fetal membrane (FM) by the end of gestation, suggesting a local role for this protein in the mechanism of parturition. However, injection of EG-VEGF to gravid mice did not induce labor, suggesting a different role for EG-VEGF in parturition. Here, we searched for its role in the FM in relation to human parturition. Human pregnant sera and total FM, chorion, and amnion were collected during the second and third trimesters from preterm no labor, term no labor, and term labor patients. Primary human chorion trophoblast and FM explants cultures were also used. We demonstrate that circulating EG-VEGF increased toward term and significantly decreased at the time of labor. EG-VEGF production was higher in the FM compared to placentas matched for gestational age. Within the FM, the chorion was the main source of EG-VEGF. EG-VEGF receptors, PROKR1 and PROKR2, were differentially expressed within the FM with increased expression toward term and an abrupt decrease with the onset of labor. In chorion trophoblast and FM explants collected from nonlaboring patients, EG-VEGF decreased metalloproteinase-2 and -9 activities and increased PGDH (prostaglandin-metabolizing enzyme) expression. Altogether these data demonstrate that EG-VEGF is a new cytokine that acts locally to ensure FM protection in late pregnancy. Its fine contribution to the initiation of human labor is exhibited by the abrupt decrease in its levels as well as a reduction in its receptors.


Assuntos
Córion/metabolismo , Regulação para Baixo , Trabalho de Parto/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adulto , Âmnio/metabolismo , Células Cultivadas , Cesárea , Córion/citologia , Feminino , Humanos , Trabalho de Parto/sangue , Placenta/metabolismo , Placentação , Gravidez , Segundo Trimestre da Gravidez , Terceiro Trimestre da Gravidez , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Peptídeos/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Técnicas de Cultura de Tecidos , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/sangue
3.
Biomed Res Int ; 2014: 309082, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25110672

RESUMO

Angiogenesis is a key process for proper placental development and for the success of pregnancy. Although numerous in vitro methods have been developed for the assessment of this process, relatively few reliable in vivo methods are available to evaluate this activity throughout gestation. Here we report an in vivo technique that specifically measures placental neovascularization. The technique is based on the measurement of a fluorescent alpha v beta 3 (αvß3) integrin-targeting molecule called Angiolone-Alexa-Fluor 700. The αvß3 integrin is highly expressed by endothelial cells during the neovascularization and by trophoblast cells during their invasion of the maternal decidua. Angiolone was injected to gravid mice at 6.5 and 11.5 days post coitus (dpc). The fluorescence was analyzed one day later at 7.5 and 12.5 dpc, respectively. We demonstrated that (i) Angiolone targets αvß3 protein in the placenta with a strong specificity, (ii) this technique is quantitative as the measurement was correlated to the increase of the placental size observed with increasing gestational age, and (iii) information on the outcome is possible, as abnormal placentation could be detected early on during gestation. In conclusion, we report the validation of a new noninvasive and quantitative method to assess the placental angiogenic activity, in vivo.


Assuntos
Troca Materno-Fetal , Neovascularização Fisiológica , Oligopeptídeos/metabolismo , Animais , Bioensaio , Feminino , Fluorescência , Imageamento Tridimensional , Camundongos , Placenta/anormalidades , Placenta/metabolismo , Gravidez
4.
Am J Physiol Endocrinol Metab ; 306(4): E443-56, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24368670

RESUMO

In utero exposure to nicotine is associated with increased risk of numerous adverse fetal and neonatal outcomes, which suggests that it acts directly to affect placental development and the establishment of the fetomaternal circulation (FC). This study used both in vivo [Wistar rats treated with 1 mg/kg nicotine from 2 wk prior to mating until gestational day (GD) 15] and in vitro (RCHO-1 cell line; treated with 10(-9) to 10(-3)M nicotine) models to examine the effects of nicotine on these pathways. At GD 15, control and treated placentas were examined for the impact of nicotine on 1) trophoblast invasion, proliferation, and degree of hypoxia, 2) labyrinth vascularization, 3) expression of key genes of placental development, and 4) expression of placental angiogenic factors. The RCHO-1 cell line was used to determine the direct effects of nicotine on trophoblast differentiation. Our in vivo experiments show that nicotine inhibits trophoblast interstitial invasion, increases placental hypoxia, downregulates labyrinth vascularization as well as key transcription factors Hand1 and GCM1, and decreases local and circulating EG-VEGF, a key placental angiogenic factor. The in vitro experiments confirmed the inhibitory effects of nicotine on the trophoblast migration, invasion, and differentiation processes and demonstrated that those effects are most likely due to a dysregulation in the expression of nicotine receptors and a decrease in MMP9 activity. Taken together, these data suggest that adverse effects of maternal smoking on pregnancy outcome are due in part to direct and endocrine effects of nicotine on the main processes of placental development and establishment of FC.


Assuntos
Nicotina/farmacologia , Placenta/efeitos dos fármacos , Placentação/efeitos dos fármacos , Trofoblastos/efeitos dos fármacos , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Feminino , Placenta/metabolismo , Gravidez , Ratos , Ratos Wistar , Trofoblastos/citologia , Trofoblastos/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
5.
Gynecol Obstet Fertil ; 41(9): 562-5, 2013 Sep.
Artigo em Francês | MEDLINE | ID: mdl-23972922

RESUMO

In spite of improvements in assisted reproductive technology (ART) during the last 30 years, the rate of pregnancy remains constrained, as only about 25 % of embryo transfer lead to successful pregnancies, even with an average of two embryos replaced. Embryo selection is currently based on the establishment of morphokinetic scores, a method that obviously exhibits limitations. Therefore, the assessment of embryo development potency by criteria of higher predictive value is mandatory in order to increase the rates of pregnancy. Nowadays, there is increasing evidence that angiogenic factors might contribute to the success of the implantation and to the pregnancy outcome. Among these factors, prokineticin 1 (PROK1) and its receptors (PROKRs) constitute new targets that showed over the last ten years strong biological features directly linked to ovarian physiology, endometrial receptivity, embryo implantation and thus successful pregnancies. In ART, the rates of circulating PROK1 were reported in 2012 as significantly linked to the quality of embryonic cohort, as well as to the rates of pregnancy. Our preliminary data suggest a high potential of this cytokine in the success of implantation and pregnancy, and strongly overtones the emergency to investigate the value of its measurement in conditioned media of oocytes and embryo cultures in ART.


Assuntos
Biomarcadores/sangue , Implantação do Embrião/fisiologia , Técnicas de Reprodução Assistida , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/sangue , Células Cultivadas , Meios de Cultivo Condicionados/química , Técnicas de Cultura Embrionária , Transferência Embrionária , Feminino , Humanos , Oócitos/metabolismo , Gravidez , Resultado do Tratamento , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/análise , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/fisiologia
6.
Pathol Biol (Paris) ; 61(5): 178-83, 2013 Oct.
Artigo em Francês | MEDLINE | ID: mdl-23647696

RESUMO

Gestational trophoblastic disease (MGT) includes a wide spectrum of pathologies of the placenta, ranging from benign precancerous lesions, with gestational trophoblastic tumors. Metastases are the leading causes of death as a result of this tumor. They represent a major problem for obstetrics and for the public health system. To date, there is no predictor of the progression of molar pregnancies to gestational trophoblastic tumor (GTT). Only an unfavorable plasma hCG monitoring after evacuation of hydatidiform mole is used to diagnose a TTG. The causes of the development of this cancer are still poorly understood. Increasing data in the literature suggests a close association between the development of this tumor and poor placental vascularization during the first trimester of pregnancy. The development of the human placenta depends on a coordination between the trophoblast and endothelial cells. A disruption in the expression of angiogenic factors could contribute to uterine or extra-uterine tissue invasion by extravillous trophoblast, contributing to the development of TTG. This review sheds lights on the phenomenon of angiogenesis during normal and abnormal placentation, especially during the MGT and reports preliminary finding concerning, the variability of expression of "Endocrine Gland-Derived Vascular Endothelial Growth Factor" (EG-VEGF), a specific placental angiogenic factor, in normal and molar placentas, and the potential role of differentiated expressions of the main placental angiogenic factors in the scalability of hydatidiform moles towards a recovery or towards the development of gestational trophoblastic tumor. Deciphering the mechanisms by which the angiogenic factor influences these processes will help understand the pathophysiology of MGT and to create opportunities for early diagnosis and treatment of the latter.


Assuntos
Doença Trofoblástica Gestacional/fisiopatologia , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/fisiologia , Gonadotropina Coriônica/sangue , Feminino , Doença Trofoblástica Gestacional/patologia , Doença Trofoblástica Gestacional/terapia , Humanos , Mola Hidatiforme/fisiopatologia , Neovascularização Patológica/fisiopatologia , Neovascularização Fisiológica/fisiologia , Placenta/irrigação sanguínea , Gravidez , Neoplasias Uterinas/fisiopatologia
7.
Cell Mol Life Sci ; 70(3): 511-25, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22941044

RESUMO

Identifiable causes of fetal growth restriction (FGR) account for 30 % of cases, but the remainders are idiopathic and are frequently associated with placental dysfunction. We have shown that the angiogenic factor endocrine gland-derived VEGF (EG-VEGF) and its receptors, prokineticin receptor 1 (PROKR1) and 2, (1) are abundantly expressed in human placenta, (2) are up-regulated by hypoxia, (3) control trophoblast invasion, and that EG-VEGF circulating levels are the highest during the first trimester of pregnancy, the period of important placental growth. These findings suggest that EG-VEGF/PROKR1 and 2 might be involved in normal and FGR placental development. To test this hypothesis, we used placental explants, primary trophoblast cultures, and placental and serum samples collected from FGR and age-matched control women. Our results show that (1) EG-VEGF increases trophoblast proliferation ([(3)H]-thymidine incorporation and Ki67-staining) via the homeobox-gene, HLX (2) the proliferative effect involves PROKR1 but not PROKR2, (3) EG-VEGF does not affect syncytium formation (measurement of syncytin 1 and 2 and ß hCG production) (4) EG-VEGF increases the vascularization of the placental villi and insures their survival, (5) EG-VEGF, PROKR1, and PROKR2 mRNA and protein levels are significantly elevated in FGR placentas, and (6) EG-VEGF circulating levels are significantly higher in FGR patients. Altogether, our results identify EG-VEGF as a new placental growth factor acting during the first trimester of pregnancy, established its mechanism of action, and provide evidence for its deregulation in FGR. We propose that EG-VEGF/PROKR1 and 2 increases occur in FGR as a compensatory mechanism to insure proper pregnancy progress.


Assuntos
Retardo do Crescimento Fetal/metabolismo , Placenta/metabolismo , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/metabolismo , Hipóxia Celular , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Feminino , Retardo do Crescimento Fetal/patologia , Células Gigantes/citologia , Proteínas de Homeodomínio/metabolismo , Humanos , Placenta/citologia , Placentação , Gravidez , Primeiro Trimestre da Gravidez , RNA Mensageiro/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Peptídeos/genética , Receptores de Peptídeos/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Fatores de Transcrição/metabolismo , Trofoblastos/citologia , Trofoblastos/metabolismo , Regulação para Cima/efeitos dos fármacos , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética
8.
Cell Mol Life Sci ; 69(9): 1537-50, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22138749

RESUMO

Endocrine gland-derived vascular endothelial growth factor (EG-VEGF) is an angiogenic factor reported to be specific for endocrine tissues, including the placenta. Its biological activity is mediated via two G protein-coupled receptors, prokineticin receptor 1 (PROKR1) and prokineticin receptor 2 (PROKR2). We have recently shown that (i) EG-VEGF expression peaks between the 8th and 11th weeks of gestation, (ii) its mRNA and protein levels are up-regulated by hypoxia, (iii) EG-VEGF is a negative regulator of trophoblast invasion and (iv) its circulating levels are increased in preeclampsia (PE), the most threatening pathology of pregnancy. Here, we investigated the regulation of the expression of EG-VEGF and its receptors by hCG, a key pregnancy hormone that is also deregulated in PE. During the first trimester of pregnancy, hCG and EG-VEGF exhibit the same pattern of expression, suggesting that EG-VEGF is potentially regulated by hCG. Both placental explants (PEX) and primary cultures of trophoblasts from the first trimester of pregnancy were used to investigate this hypothesis. Our results show that (i) LHCGR, the hCG receptor, is expressed both in cyto- and syncytiotrophoblasts, (ii) hCG increases EG-VEGF, PROKR1 and PROKR2 mRNA and protein expression in a dose- and time-dependent manner, (iii) hCG increases the release of EG-VEGF from PEX conditioned media, (iv) hCG effects are transcriptional and post-transcriptional and (v) the hCG effects are mediated by cAMP via cAMP response elements present in the EG-VEGF promoter region. Altogether, these results demonstrate a new role for hCG in the regulation of EG-VEGF and its receptors, an emerging regulatory system in placental development.


Assuntos
Gonadotropina Coriônica/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Peptídeos/metabolismo , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/metabolismo , Sequência de Bases , Células Cultivadas , Gonadotropina Coriônica/farmacologia , Primers do DNA/genética , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Técnicas In Vitro , Modelos Biológicos , Dados de Sequência Molecular , Placenta/efeitos dos fármacos , Placenta/metabolismo , Placentação , Gravidez , Primeiro Trimestre da Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores do LH/metabolismo , Receptores de Peptídeos/genética , Trofoblastos/efeitos dos fármacos , Trofoblastos/metabolismo , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/genética
9.
Endocrine ; 36(2): 281-90, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19693712

RESUMO

We have previously shown that in utero nicotine exposure causes impaired fertility, follicle immaturity, and ovarian dysfunction in adult female rat offspring. These characteristics overtly resemble the clinical profile of polycystic ovarian syndrome (PCOS) and recent studies have shown that thiazolidinediones such as rosiglitazone improve fertility in women with PCOS but the mechanism is not well defined. Our goal was to examine whether rosiglitazone would (1) ameliorate the altered ovarian physiology that occurs following fetal and neonatal exposure to nicotine and (2) to examine whether this could be due to normalization of ovarian vascularization. At weaning, offspring of nicotine-exposed dams were given either vehicle (NV) or rosiglitazone (3 mg kg(-1) day(-1); NR). Offspring of saline-exposed dams received vehicle (SV). Tissues were collected when the female offspring reached 26 weeks of age. NV animals had reduced granulosa cell proliferation and increased ovarian cell apoptosis. Treatment with rosiglitazone increased proliferation, and decreased apoptosis, compared NV animals. NV animals had decreased ovarian vascularity relative to controls, whereas NR animals had an intermediate level of ovarian vessel density. Moreover, ovaries from NV animals had decreased levels of the pro-angiogenic growth factors vascular endothelial growth factor (VEGF) and endocrine gland-derived VEGF both of which were increased with rosiglitazone treatment. Rosiglitazone reversed some of the nicotine effects in the ovary and increased ovarian vascularization, follicle maturation and improved oocyte competence. Rosiglitazone may be an important treatment option for PCOS and the present study provides a potential mechanism by which rosiglitazone may have beneficial effects on fertility in these patients.


Assuntos
Fertilidade/efeitos dos fármacos , Nicotina/efeitos adversos , Ovário/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Tiazolidinedionas/farmacologia , Animais , Animais Recém-Nascidos , Vasos Sanguíneos/efeitos dos fármacos , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Feminino , Fertilidade/fisiologia , Hipoglicemiantes/farmacologia , Infertilidade Feminina/fisiopatologia , Exposição Materna/efeitos adversos , Ovário/irrigação sanguínea , Ovário/fisiologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Ratos , Ratos Wistar , Rosiglitazona
10.
Placenta ; 28(10): 1049-58, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17531315

RESUMO

Compelling evidence indicates that vascular endothelial growth factor (VEGF) is an important mediator of placental angiogenesis and appears to be disregulated in pre-eclampsia (PE). Recently, we characterised the expression of EG-VEGF (endocrine gland-derived vascular endothelial growth factor), also known as prokineticin 1 (PK1) in human placenta during the first trimester of pregnancy and showed that this factor is likely to play an important role in human placentation. However, because it is impossible to prospectively study placentation in humans, it has been impossible to further characterise EG-VEGF expression throughout complete gestation and especially at critical gestational ages for PE development. In the present study, we used mouse placenta to further characterise EG-VEGF expression throughout gestation. We investigated the pattern of expression of EG-VEGF and its receptors, PKR1 and PKR2 at the mRNA and protein levels. Our results show that EG-VEGF and VEGF exhibit different patterns of expression and different localisations in the mouse placenta. EG-VEGF was mainly localised in the labyrinth whereas VEGF was mainly present in glycogen and giant cells. EG-VEGF mRNA and protein levels were highest before 10.5days post coitus (dpc) whereas those of VEGF showed stable expression throughout gestation. PKR1 protein was localised to the labyrinth layer and showed the same pattern of expression as EG-VEGF whereas PKR2 expression was maintained over 10.5dpc with both trophoblastic and endothelial cell localisations. Altogether these findings suggest that EG-VEGF may have a direct effect on both endothelial and trophoblastic cells and is likely to play an important role in mouse placentation.


Assuntos
Placenta/metabolismo , Receptores Acoplados a Proteínas G/biossíntese , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/biossíntese , Animais , Feminino , Hormônios Gastrointestinais/biossíntese , Idade Gestacional , Imuno-Histoquímica , Camundongos , Neuropeptídeos/biossíntese , Gravidez , RNA Mensageiro/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese
11.
Placenta ; 28(8-9): 907-11, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17254632

RESUMO

Copper is an essential trace element for successful pregnancy. However, the mechanisms by which copper is transported from maternal circulation to the fetus have not been clearly elucidated. Two proteins, cellular prion (PrP(C)) and COMMD1, are known to be responsible for prion diseases and canine copper toxicosis, respectively, and are thought to play a role in copper homeostasis. However, their placental expression and localization throughout human gestation are still unknown. In this study, we used quantitative RT-PCR, western blotting and immunohistochemistry to investigate in detail the expression and localization of PrP(C) and COMMD1 proteins in human placenta throughout pregnancy. Our results show that both proteins are expressed in human placenta. PrP(C) showed the highest mRNA and protein expression levels during the first trimester of pregnancy. PrP(C) and COMMD1 proteins are similarly localized within the placental villi. Both proteins are present in the syncytiotrophoblast, the cytotrophoblast, vascular endothelial cells and Hofbauer cells. These data offer some insights into possible roles for PrP(C) and COMMD1 within the placenta.


Assuntos
Placenta , Trofoblastos , Animais , Vilosidades Coriônicas/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Placenta/metabolismo , Gravidez , Primeiro Trimestre da Gravidez , Príons , RNA Mensageiro/metabolismo , Trofoblastos/metabolismo
12.
J Endocrinol ; 176(2): 175-84, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12553866

RESUMO

In the late-gestation sheep, increased fetal plasma cortisol concentration and placental oestradiol (E(2)) output contribute to fetal organ maturation, in addition to the onset of parturition. Both cortisol and E(2) are believed to regulate the enzyme 11beta-hydroxysteroid dehydrogenase type 1 (11beta-HSD1), which interconverts bioactive 11-hydroxy glucocorticoids and their inactive 11-keto metabolites. 11beta-HSD1, abundantly expressed in fetal liver, operates primarily as a reductase enzyme to produce bioactive cortisol and thus regulates local hepatic glucocorticoid concentrations. Cortisol acts through the glucocorticoid receptor (GR) present in the liver. In this study, we examined the effects of cortisol and E(2) on hepatic 11beta-HSD1 and GR in the liver of chronically catheterized sheep fetuses treated with saline (n=5), cortisol (1.35 mg/h; n=5), saline+4-hydroxyandrostendione, a P450 aromatase inhibitor (4-OHA; 1.44 mg/h; n=5), or cortisol+4-OHA (n=5). Cortisol infusion resulted in increased plasma concentrations of fetal cortisol and E(2); concurrent administration of 4-OHA attenuated the increase in plasma E(2) concentrations. Using immunohistochemistry, we showed that fetal hepatocytes expressed both 11beta-HSD1 and GR proteins. Cortisol treatment increased GR in both cytosol and nuclei of hepatocytes; concurrent administration of 4-OHA was associated with distinct nuclear GR staining. Western blot revealed that cortisol, in the absence of increased E(2) concentrations, significantly increased concentrations of 11beta-HSD1 (34 kDa) and GR (95 kDa) proteins. 11beta-HSD1 enzyme activity was measured in the liver microsomal fraction in the presence of [(3)H]cortisone (10(-)(6) M) or [(3)H]cortisol (10(-)(6) M) and NADPH (reductase activity) or NADP(+) (dehydrogenase activity) respectively. 11beta-HSD1 reductase activity was significantly greater in the presence of cortisol. In summary, we found that, in sheep during late gestation, cortisol increased both 11beta-HSD1 and GR in the fetal liver, and these effects were accentuated in the absence of increased E(2).


Assuntos
Androstenodiona/análogos & derivados , Estradiol/farmacologia , Hidrocortisona/farmacologia , Hidroxiesteroide Desidrogenases/metabolismo , Fígado/embriologia , Fígado/metabolismo , Receptores de Glucocorticoides/metabolismo , 11-beta-Hidroxiesteroide Desidrogenases , Análise de Variância , Androstenodiona/farmacologia , Animais , Inibidores da Aromatase , Western Blotting/métodos , Inibidores Enzimáticos/farmacologia , Feminino , Idade Gestacional , Hidroxiesteroide Desidrogenases/análise , Imuno-Histoquímica/métodos , Fígado/efeitos dos fármacos , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Gravidez , Receptores de Glucocorticoides/análise , Ovinos , Estimulação Química
13.
J Clin Endocrinol Metab ; 87(11): 5274-82, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12414902

RESUMO

Prostaglandins (PGs) act as potent uterotonins at the time of labor. Prostaglandin E synthase (PGES) is responsible for the formation of PGE(2), a uterotonin. PGI(2) is synthesized by the prostaglandin I synthase enzyme (PGIS) and contributes to relaxation in the lower uterine segment. We examined the expression of membrane-bound PGES and PGIS in myometrium from pregnant women during preterm and term labor. Tissues were collected from the lower uterine segment from preterm no labor, preterm labor, term no labor, and term labor patients and used for immunohistochemistry and Western blot analysis using specific antibodies. Immunoreactive (ir-) PGES and PGIS proteins were localized to the cytoplasm of myocytes of the myometrium and vascular smooth muscle cells. Ir-PGES was also detected in vascular endothelial cells. Western blot analyses revealed a predominant protein band of 180 kDa, and a second 16-kDa band for ir-PGES and 56-kDa band for ir-PGIS. There was no significant change in ir-PGES protein (180 or 16 kDa) or mRNA levels with preterm or term labor or gestational age. There was a significant decrease in PGIS mRNA and protein with advancing gestational age. We conclude that the gestational age decrease in the inhibitory PGIS is consistent with lessening of its influence in myometrium at the time of labor. The lack of change in PGES indicates that alterations at other points along the pathway of arachidonic acid metabolism may be of greater importance in affecting local changes in PGE(2).


Assuntos
Sistema Enzimático do Citocromo P-450/genética , Expressão Gênica , Oxirredutases Intramoleculares/genética , Trabalho de Parto/fisiologia , Miométrio/metabolismo , Actinas/genética , Western Blotting , Sistema Enzimático do Citocromo P-450/análise , Sistema Enzimático do Citocromo P-450/metabolismo , Citoplasma/enzimologia , Endotélio Vascular/enzimologia , Feminino , Imunofluorescência , Corantes Fluorescentes , Idade Gestacional , Humanos , Imuno-Histoquímica , Hibridização In Situ , Oxirredutases Intramoleculares/análise , Oxirredutases Intramoleculares/metabolismo , Músculo Liso Vascular/enzimologia , Miométrio/irrigação sanguínea , Gravidez , Prostaglandina-E Sintases , RNA Mensageiro/análise
14.
J Clin Endocrinol Metab ; 87(10): 4797-805, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12364476

RESUMO

Preeclampsia (PE) is a major cause of maternal and perinatal morbidity and mortality. The genesis of PE is related to deficient trophoblast invasion of maternal spiral arteries, which might result in a reduction of placental (PL) oxygen (O(2)). An absence of increased O(2) that normally occurs around the 10-12th wk of gestation results in aberrant expression of genes that might contribute to the pathophysiology of PE. We examined the expression and regulation of PL 11 beta-hydroxysteroid dehydrogenase 2 (11 beta-HSD) in normal pregnancies and in PE. Two types of 11 beta-HSD exist in the placenta, 11 beta-HSD1 and 11 beta-HSD2. 11 beta-HSD2 is thought to protect the fetus from cortisol excess. In PE, both the expression and activity of PL 11 beta-HSD2 were reduced significantly compared with those in age-matched controls. As PE is associated with a reduction of PL O(2), we next investigated whether in normal pregnancy 11 beta-HSD2 expression changes at the time of the increase in O(2). 11 beta-HSD2 was detected as early as 5 wk, with expression limited to the syncytiotrophoblast (ST). At 10-12 wk, this expression increased and was also found in the cytotrophoblast and extravillous trophoblast. These results were substantiated by Western blot. The ability of O(2) to regulate 11 beta-HSD2 was determined both in cultures of villous explant from early gestation and in term trophoblast cells after incubation under 3% or 20% O(2). Villous explants cultured under 20% O(2) showed higher enzyme activity and expression compared with 3% O(2). Term trophoblast cells also exhibited higher enzyme activity at 20% vs. 3% O(2). No change in 11 beta-HSD1 expression was observed in early pregnancy or in PE. This is the first report to suggest that 11 beta-HSD2 is O(2) dependent in first and third trimester placenta during human gestation.


Assuntos
Hidroxiesteroide Desidrogenases/análise , Oxigênio/farmacologia , Placenta/enzimologia , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2 , Western Blotting , Células Cultivadas , Feminino , Idade Gestacional , Humanos , Hidroxiesteroide Desidrogenases/metabolismo , Imuno-Histoquímica , Oxigênio/administração & dosagem , Pré-Eclâmpsia/enzimologia , Gravidez , Distribuição Tecidual , Trofoblastos/enzimologia
15.
Mol Cell Endocrinol ; 185(1-2): 135-44, 2001 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-11738803

RESUMO

A general characteristic of fetal endocrine maturation across different species is the enhanced activity of the fetal hypothalamic-pituitary-adrenal (HPA) axis during late gestation. Precocious activation of this axis may occur when the fetus is exposed to an adverse intra-uterine environment, such as hypoxemia. HPA development is associated with increased levels of ACTH(1-39) and adrenal corticosteroids (cortisol in sheep and human) in the fetal circulation, and increased expression of mRNA encoding corticotrophin releasing hormone (CRH) in the hypothalamus, proopiomelanocortin (POMC) in the pituitary, and key steroidogenic enzymes in the fetal adrenal. At term, increased levels of cortisol act on the placenta/trophoblast derived cells to increase expression of prostaglandin synthase Type II (PGHS-II). In human gestation, cortisol also decreases expression of 15-hydroxyprostaglandin dehydrogenase (PGDH) in chorionic trophoblast cells. Increased synthesis and decreased metabolism of prostaglandin (PG) results, during late gestation, in enhanced output of primary PG, which in turn increases the activity of 11 beta-hydroxysteroid dehydrogenase (11 beta HSD) in the human fetal membranes. Increased chorionic 11 beta HSD-1 results in increased local generation of cortisol from cortisone, with further paracrine/autocrine stimulation of PG output. Increased fetal cortisol contributes to the maturation of organ systems required for postnatal extra-uterine survival. However, excessive levels of feto-placental glucocorticoid, derived from maternal administration of synthetic corticosteroids or sustained endogenous fetal cortisol production, results in intrauterine growth restriction. Fetal sheep, exposed to maternal betamethasone in late gestation, develop insulin resistance and exaggerated adrenal responses to HPA stimulation by 6-12 months postnatal life. Thus, the level of fetal HPA activity is crucial not only for determining gestation length, but may also predict pathophysiologic adjustments in later life.


Assuntos
Sistema Hipotálamo-Hipofisário/embriologia , Sistema Hipófise-Suprarrenal/embriologia , Animais , Desenvolvimento Embrionário e Fetal/fisiologia , Feminino , Glucocorticoides/sangue , Humanos , Sistema Hipotálamo-Hipofisário/crescimento & desenvolvimento , Parto/metabolismo , Sistema Hipófise-Suprarrenal/crescimento & desenvolvimento , Placenta/embriologia , Gravidez
16.
J Clin Endocrinol Metab ; 86(11): 5585-92, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11701739

RESUMO

In human pregnancy, cortisol and PGs are involved in the onset of labor and play an important role in the mechanisms leading to parturition. Recent studies have shown that at term, cortisol increases PG synthesis and decreases PG metabolism in chorion trophoblast (CT) cells. In CT, 11 beta-hydroxysteroid oxidase type 1 (11 beta-HSD1) converts biologically inactive cortisone to cortisol to regulate cortisol availability. In the present study, we have investigated whether 11 beta-HSD1 activity could be influenced by PGs. We have shown that in CT, PGF2alpha rapidly increased 11 beta-HSD1 reductase activity in a dose-dependent manner via the PGF2alpha receptor, localized in the fetal membranes. PGF2alpha stimulated 11 beta-HSD1 activity through increased intracellular calcium mobilization, activation of PKC, and the phosphorylation of the 11 beta-HSD enzyme. We propose that within CT there is a novel feed forward loop by which PGF2alpha acts to promote cortisol production from cortisone through increases in 11beta-HSD1, and this in turn leads to further net PG output for the onset of labor and birth.


Assuntos
Dinoprosta/farmacologia , Hidrocortisona/biossíntese , Hidroxiesteroide Desidrogenases/biossíntese , Trabalho de Parto/fisiologia , 11-beta-Hidroxiesteroide Desidrogenases , Adulto , Cálcio/metabolismo , Células Cultivadas , Retroalimentação/fisiologia , Feminino , Feto/metabolismo , Corantes Fluorescentes , Fura-2 , Humanos , Imuno-Histoquímica , Luteolíticos/farmacologia , Membranas/metabolismo , Testes de Precipitina , Gravidez , Prostaglandinas F Sintéticas/farmacologia , Receptores de Prostaglandina/metabolismo , Estimulação Química , Trofoblastos/metabolismo
17.
Ann N Y Acad Sci ; 943: 225-34, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11594542

RESUMO

Increased uterine contractility at term and preterm results from activation and then stimulation of the myometrium. Activation can be provoked by mechanical stretch of the uterus and by an endocrine pathway resulting from increased activity of the fetal hypothalamic-pituitary-adrenal (HPA) axis. In fetal sheep, increased cortisol output during pregnancy regulates prostaglandin H synthase type 2 (PGHS2) expression in the placenta in an estrogen-independent manner, resulting in increased levels of PGE2 in the fetal circulation. Later increases in maternal uterine expresssion of PGHS2 require elevations of estrogen and lead to increased concentrations of PGF2alpha in the maternal circulation. Thus, regulation of PGHS2 at term is differentially controlled in fetal (trophoblast) and maternal (uterine epithelium) tissue. This difference may reflect expression of the glucocorticoid receptor (GR), but not estrogen receptor (ER), in placental trophoblast cells. In women, cortisol also contributes to increased PG production in fetal tissues through upregulation of PGHS2 (amnion and chorion) and downregulation of 15-OH PG dehydrogenase (chorion trophoblasts). The effect of cortisol on chorion expression of PGDH reverses a tonic stimulatory effect of progesterone, potentially through a paracrine or autocrine action. We have interpreted this interaction as a reflection of "progesterone withdrawal" in the primate, in relation to birth. Other agents, such as proinflammatory cytokines, similarly upregulate PGHS2 and decrease expression of PGDH, indicating the presence of several mechanisms by which labor at term or preterm may be initiated. These different mechanisms need to be considered in the development of strategies for the detection and management of the patient in preterm labor.


Assuntos
Trabalho de Parto Prematuro/fisiopatologia , Hormônio Liberador da Corticotropina/fisiologia , Feminino , Humanos , Trabalho de Parto/fisiologia , Gravidez , Prostaglandinas/metabolismo , Prostaglandinas/fisiologia
18.
Biol Reprod ; 64(4): 1019-32, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11259246

RESUMO

Birth in many animal species and in humans is associated with activation of hypothalamic-pituitary-adrenal function in the fetus and the increased influence of glucocorticoids on trophoblast cells of the placenta and fetal membranes. We suggest that in ovine pregnancy glucocorticoids directly increase fetal placental prostaglandin production, and indirectly increase prostaglandin production by maternal uterine tissues through the stimulation of placental estradiol synthesis. The events of ovine parturition are compared with those of human parturition. In the latter, we suggest similar direct effects of glucocorticoids on prostaglandin synthesis and metabolism in fetal membranes and similar indirect effects mediated by glucocorticoid-stimulated increases in intrauterine corticotropin-releasing hormone expression.


Assuntos
Glândulas Suprarrenais/embriologia , Glucocorticoides/fisiologia , Hipotálamo/embriologia , Trabalho de Parto/fisiologia , Hipófise/embriologia , Prostaglandinas/biossíntese , Glândulas Suprarrenais/fisiologia , Animais , Hormônio Liberador da Corticotropina/fisiologia , Feminino , Glucocorticoides/farmacologia , Humanos , Hipotálamo/fisiologia , Hipófise/fisiologia , Placenta/efeitos dos fármacos , Placenta/metabolismo , Gravidez , Prostaglandinas/metabolismo , Ovinos , Útero/metabolismo
19.
Endocr Res ; 26(4): 489-504, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11196419

RESUMO

Birth in most animal species is triggered by the fetus through activation of the fetal hypothalamic-pituitary-adrenal (HPA) axis. Preterm birth, may be associated with precocious activation of fetal HPA function, reflecting the fetal response to an adverse intrauterine environment. There is a progressive and concurrent increase of ACTH1-39 and cortisol (F) in the circulation of fetal sheep during the last 15-20 days of pregnancy (term, day 145-150) associated with increased expression of hypothalamic CRH pituitary POMC and adrenal ACTH receptor and steroidogenic enzymes, particularly P450 C17. Similar changes occur with fetal hypoxemia. Negative feedback is ameliorated by decreased pituitary and hypothalamic glucocorticoid receptor, increased CBG, and altered fetal pituitary 11B-hydroxysteroid dehydrogenase type 1. Repeated fetal hypoxemia, diminishes the fetal-pituitary ACTH response, but increases fetal adrenal responsiveness. Fetuses exposed to maternal glucocorticoid in late gestation are growth restricted with altered postnatal HPA responsiveness and glycemic responses that reproduce the insulin resistance of type 2 diabetes. We conclude that the level of fetal HPA activity is crucial not only for determining gestation length, but also predicts pathophysiologic adjustment in later life.


Assuntos
Doenças dos Animais/etiologia , Animais Recém-Nascidos/fisiologia , Feto/fisiologia , Sistema Hipotálamo-Hipofisário/embriologia , Doenças do Recém-Nascido/etiologia , Trabalho de Parto/fisiologia , Sistema Hipófise-Suprarrenal/embriologia , Animais , Desenvolvimento Embrionário e Fetal , Feminino , Feto/efeitos dos fármacos , Glucocorticoides/farmacologia , Humanos , Recém-Nascido , Gravidez , Fatores de Tempo
20.
J Clin Invest ; 100(10): 2437-42, 1997 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-9366557

RESUMO

Arginine vasopressin (AVP) and corticosteroid hormones are involved in sodium reabsorption regulation in the renal collecting duct. Synergy between AVP and aldosterone has been well documented, although its mechanism remains unclear. Both aldosterone and glucocorticoid hormones bind to the mineralocorticoid receptor (MR), and mineralocorticoid selectivity depends on the MR-protecting enzyme 11 beta hydroxysteroid deshydrogenase (11-HSD), which metabolizes glucocorticoids into derivatives with low affinity for MR. We have investigated whether the activity of 11-HSD could be influenced by AVP and corticosteroid hormones. This study shows that in isolated rat renal collecting ducts, AVP increases 11-HSD catalytic activity. This effect is maximal at 10(-8) M AVP (a concentration clearly above the normal physiological range of AVP concentrations) and involves the V2 receptor pathway, while activation of protein kinase C or changes in intracellular calcium are ineffective. The stimulatory effect of AVP on 11-HSD is largely reduced after adrenalectomy, and is selectively restored by infusion of aldosterone, not glucocorticoids. We conclude that this synergy between AVP and aldosterone in controlling the activity of 11-HSD is likely to play a pivotal role in resetting mineralocorticoid selectivity, and hence sodium reabsorption capacities of the renal collecting duct.


Assuntos
Aldosterona/farmacologia , Arginina Vasopressina/farmacologia , Corticosterona/metabolismo , Hidroxiesteroide Desidrogenases/metabolismo , Córtex Renal/enzimologia , Túbulos Renais Coletores/fisiologia , Receptores de Vasopressinas/fisiologia , 1-Metil-3-Isobutilxantina/farmacologia , 11-beta-Hidroxiesteroide Desidrogenases , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Adrenalectomia , Animais , Corticosterona/farmacologia , Desamino Arginina Vasopressina/farmacologia , Dexametasona/farmacologia , Túbulos Renais Coletores/efeitos dos fármacos , Cinética , Masculino , Proteína Quinase C/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Vasopressinas/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...