Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Reprod Immunol ; 67(2): 101-11, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21999474

RESUMO

PROBLEM: The regulatory mechanisms involved in VEGF-C secretion by trophoblasts during placentation are poorly understood. We investigated whether or not decidual natural killer cell conditioned medium (dNK-CM) stimulated VEGF-C secretion in the extravillous cytotrophoblast (EVT) cell line HTR8/SVneo. METHOD OF STUDY: The effects of dNK-CM and recombinant IFN-γ on VEGF-C induction by HTR8/SVneo were studied in the absence or presence of IFN-γ or its receptor blocking antibodies, p38 inhibitor (SB202190), JAK inhibitor (JAK inhibitor-1, JI-1), and on STAT1 knockdown HTR8/SVneo. VEGF-C was quantified by ELISA. FACS was used to investigate the phosphorylations of Tyr701 or Ser727 of STAT1 on stimulated HTR8/SVneo. RESULTS: dNK-CM facilitated VEGF-C secretion by HTR8/SVneo. IFN-γ and IFN-γR1 or IFN-γR2 blocking antibodies reduced both dNK-CM- and IFN-γ-induced VEGF-C secretion. Phosphorylations on Tyr701 or Ser727 of STAT1 were elevated upon stimulation. Secretion of VEGF-C was reduced by treatment with SB202190, JI-1, or STAT1 knockdown by siRNA. CONCLUSION: VEGF-C production by trophoblasts is regulated by soluble factors secreted by dNK through p38 and JAK-STAT1 pathways.


Assuntos
Meios de Cultivo Condicionados/farmacologia , Decídua/citologia , Células Matadoras Naturais/metabolismo , Trofoblastos/metabolismo , Fator C de Crescimento do Endotélio Vascular/metabolismo , Anticorpos Bloqueadores , Linhagem Celular , Feminino , Humanos , Imidazóis/farmacologia , Interferon gama/imunologia , Interferon gama/farmacologia , Janus Quinases/antagonistas & inibidores , Janus Quinases/metabolismo , Placenta/metabolismo , Placentação , Gravidez , Piridinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno , Receptores de Interferon/antagonistas & inibidores , Receptores de Interferon/imunologia , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Receptor de Interferon gama
2.
Reprod Biol Endocrinol ; 7: 81, 2009 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-19656390

RESUMO

Endometrial carcinoma is the most common neoplasm of the female genital tract, accounting for nearly one half of all gynecologic cancers in the Western world. Although intensive research on pathological phenomena of endometrial cancer is currently going on, but exact cause and biological aspects of this disease are not well described yet. In addition to well-documented roles of gonadotropin-releasing hormone (GnRH) in hypopituitary ovarian (HPO) axis, the agonistic or antagonistic analogs (or both) of GnRH have been shown to inhibit the proliferation of a variety of human gynecologic cancers. Thus, in the present study, we further examined the possibility that GnRH induces integrin beta3 and activation of focal adhesion kinase (FAK) through mitogen-activated protein kinases (MAPKs), ERK1/2 and p38, to inhibit the growth of HEC1A endometrial cancer cell line. As a result, both GnRH-I and GnRH-II resulted in a significant increase in integrin beta3 expression and evoked the activation of FAK in a time-dependent manner in these cells. In addition, these analogs induced an activation of ERK1/2 and p38 MAPK in a time-dependent manner as downstream pathways of FAK. It appears that GnRH-II has much greater effect on the activation of FAK, ERK1/2 and p38 compared to GnRH-I in these cells. Further, we demonstrated that the growth inhibition of HEC1A cells by GnRH-I or GnRH-II is involved in the activation of integrin-FAK and ERK1/2 and p38 MAPK pathways. Taken together, these results suggest that GnRH may be involved in the inhibition of endometrial cancer cell growth via activation of integrin beta3 and FAK as a direct effect. This knowledge could contribute to a better understanding of the mechanisms implicated in the therapeutic action of GnRH and its biomedical application for the treatment against endometrial cancer.


Assuntos
Proliferação de Células/efeitos dos fármacos , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Hormônio Liberador de Gonadotropina/farmacologia , Integrina beta3/metabolismo , Western Blotting , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Neoplasias do Endométrio/metabolismo , Neoplasias do Endométrio/patologia , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Feminino , Hormônio Liberador de Gonadotropina/análogos & derivados , Humanos , Imidazóis/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Precursores de Proteínas/farmacologia , Piridinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
3.
Cancer Res ; 69(10): 4202-8, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19366794

RESUMO

Gonadotropin-releasing hormone type II (GnRH-II) has an antiproliferative effect on human endometrial cancer cells. Apoptosis in cancer cells may play a critical role in regulating cell proliferation. However, more studies are necessary to elucidate the underlying molecular mechanisms and develop potential applications of GnRH-II. Therefore, we explored the mechanisms of GnRH-II-induced apoptosis and the effects of GnRH-II on GADD45alpha activation in human endometrial cancer cell lines. GnRH-II decreased cell viability in a dose- and time-dependent manner. Apoptosis was induced with increased terminal deoxyribonucleotidyl transferase-mediated dUTP nick end labeling apoptotic cells after GnRH-II treatment. Knockdown of the endogenous GnRH-I receptor with small interfering RNA (siRNA) rescued the cells from GnRH-II-mediated cell growth inhibition and abolished the induction of apoptosis. GnRH-II activated extracellular signal-regulated kinase (ERK)-1/2 and p38 mitogen-activated protein kinase (MAPK) in a time-dependent manner, and the activation was abolished by GnRH-I receptor siRNA and MAPK inhibitors. Cells pretreated with MAPK inhibitors were rescued from GnRH-II-mediated cell growth inhibition. Moreover, both inhibitors abolished GnRH-II-induced apoptosis. GnRH-II induced GADD45alpha expression, which was abolished by knockdown of endogenous GnRH-I receptors and MAPK inhibitors. GnRH-II-stimulated cell growth inhibition was rescued by knockdown of endogenous GADD45alpha with siRNA. Cells treated with GADD45alpha siRNA were refractory to GnRH-II-induced apoptosis. Thus, GnRH-II inhibits cell growth by inducing apoptosis through binding of the GnRH-I receptor, activation of the ERK1/2 and p38 MAPK pathways, and induction of GADD45alpha signaling. This finding may provide a new concept relating to the mechanism of GnRH-II-induced antiproliferation and apoptosis in endometrial cancer cells, indicating the possibility of GnRH-II as a promising therapeutic intervention for human endometrial cancer.


Assuntos
Apoptose/fisiologia , Neoplasias do Endométrio/patologia , Hormônio Liberador de Gonadotropina/análogos & derivados , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Neoplasias do Endométrio/enzimologia , Neoplasias do Endométrio/genética , Feminino , Hormônio Liberador de Gonadotropina/farmacologia , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , RNA Interferente Pequeno/genética , Receptores LHRH/deficiência , Receptores LHRH/genética , Receptores LHRH/fisiologia , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
4.
Mol Endocrinol ; 23(7): 1014-21, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19372239

RESUMO

The potential roles of GnRH I and GnRH II have been assigned in promoting the invasive capacity of human trophoblasts by regulating matrix metalloproteinases-2 and -9, type I tissue inhibitor of matrix metalloproteinase, and urokinase plasminogen activator/plasminogen activator inhibitor protease systems during human placentation, and GnRH II has been shown to be more potent than GnRH I. However, the mechanisms for the differential effects of these two hormones remain unclear. In this study, we examined the invasion-promoting effects and the signaling pathways of GnRH I and GnRH II in human trophoblasts. The data revealed that both GnRH I and GnRH II were key autocrine and/or paracrine regulators in facilitating trophoblast invasion. The GnRH receptor antagonist (Antide) and specific small interfering RNA for GnRH receptor inhibited the regulatory effects of GnRH I, but not GnRH II, on trophoblast invasion. Both GnRH I and II activated protein kinase C, ERK1/2, and c-Jun N-terminal kinase to mediate their effects on trophoblast invasion, whereas only GnRH II elicited invasion-promoting action through transactivating the tyrosine kinase activity of epidermal growth factor receptor in trophoblasts. Our observations elucidate a ligand-dependent selective cross-communication between GnRH receptor and epidermal growth factor receptor signaling systems in human trophoblastic cell, and this would further our understanding on the differentially biological significance of these two forms of GnRH in extrapituitary tissues.


Assuntos
Movimento Celular/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/análogos & derivados , Precursores de Proteínas/farmacologia , Trofoblastos/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Receptores ErbB/fisiologia , Hormônio Liberador de Gonadotropina/farmacologia , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 2 da Matriz/fisiologia , Metaloproteinase 9 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , Modelos Biológicos , Oligopeptídeos/farmacologia , RNA Interferente Pequeno/farmacologia , Receptores LHRH/antagonistas & inibidores , Receptores LHRH/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Trofoblastos/fisiologia
5.
Reproduction ; 137(5): 769-77, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19208750

RESUMO

Type I GnRH (GnRH-I, GNRH1) and type II GnRH (GnRH-II, GNRH2), each encoded by separate genes, have been identified in humans. The tissue distribution and functional regulation of GnRH-I and GnRH-II clearly differ despite their comparable cDNA and genomic structures. These hormones exert their effects by binding to cell surface transmembrane G protein coupled receptors and stimulating the Gq/11 subfamily of G proteins. The hypothalamus and pituitary are the main origin and target sites of GnRH, but numerous studies have demonstrated that extra-hypothalamic GnRH and extra-pituitary GnRH receptors exist in different reproductive tissues such as the ovary, endometrium, placenta, and endometrial cancer cells. In addition to endocrine regulation, GnRH is also known to act in an autocrine and paracrine manner to suppress cell proliferation and activate apoptosis in the endometrium and endometrial cancer cells through several mechanisms. Both GnRH-I and GnRH-II exhibit regulatory roles in tissue remodelling during embryo implantation and placentation, which suggests that these hormones may have important roles in embryo implantation and early pregnancy. The presence of varied GnRH and GnRH receptor systems demonstrate their different roles in distinct tissues using dissimilar mechanisms. These may result in the generation of new GnRH analogues used for several hormone-related diseases.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Transdução de Sinais , Útero/metabolismo , Animais , Apoptose , Comunicação Autócrina , Proliferação de Células , Feminino , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/uso terapêutico , Humanos , Comunicação Parácrina , Receptores LHRH/metabolismo , Doenças Uterinas/tratamento farmacológico , Doenças Uterinas/metabolismo , Útero/patologia
6.
Mol Hum Reprod ; 14(5): 281-9, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18430759

RESUMO

Pre-eclampsia is often associated with inadequate cytotrophoblast invasion and remodelling of the uterine spiral arteries. Examining a first trimester, 2D in vitro explant culture model which mimics in vivo placentation, including trophoblast column formation and extravillous cytotrophoblast (EVT) migration, we previously suggested that excessive maternal decidual natural killer cell interferon (IFN-gamma) limits EVT migration. Types-1 and -2 insulin-like growth factor (IGF-1, IGF-2) are trophic for EVT, act through their surface receptors, IGFR-1 and IGFR-2, and are regulated by the IGF-binding proteins (IGFBPs). Could the observed IFN-gamma-mediated inhibition of EVT outgrowth and migration be related to either expression changes of IGF-1 or IGF-2, their receptors, their binding proteins, or apoptosis? Using the 2D explant culture model, we examined the effect of IFN-gamma exposure on IGF-1 and -2, IGFR-1 and -2, IGFBPs and apoptosis. IFN-gamma relatively increased IGF-1 and -2 secretion. In EVT, IFN-gamma decreased IGFR-2, but not IGFR-1 expression. IGBP-2, -3 and -4 production were not influenced by IFN-gamma. IFN-gamma induced trophoblast apoptosis measured by the highly sensitive M30 neo-epitope, but not caspase 3 activity, in conditioned medium and EVT cell lysates. The observed IFN-gamma-mediated EVT migration inhibition may occur through the down-regulation of IGFR-2 and subtle induction of EVT apoptosis.


Assuntos
Vilosidades Coriônicas/efeitos dos fármacos , Interferon gama/farmacologia , Primeiro Trimestre da Gravidez/efeitos dos fármacos , Somatomedinas/fisiologia , Trofoblastos/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Apoptose/genética , Movimento Celular/efeitos dos fármacos , Extensões da Superfície Celular/efeitos dos fármacos , Extensões da Superfície Celular/fisiologia , Células Cultivadas , Vilosidades Coriônicas/metabolismo , Vilosidades Coriônicas/fisiologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Feminino , Humanos , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/genética , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/metabolismo , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Fator de Crescimento Insulin-Like I/fisiologia , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/metabolismo , Fator de Crescimento Insulin-Like II/fisiologia , Integrinas/genética , Integrinas/metabolismo , Técnicas de Cultura de Órgãos , Gravidez , Primeiro Trimestre da Gravidez/genética , Primeiro Trimestre da Gravidez/metabolismo , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Receptor IGF Tipo 2/genética , Receptor IGF Tipo 2/metabolismo , Somatomedinas/genética , Somatomedinas/metabolismo , Trofoblastos/metabolismo , Trofoblastos/fisiologia
7.
J Immunol ; 177(12): 8522-30, 2006 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17142750

RESUMO

Abnormal placentation results in either inadequate (consequences: recurrent miscarriage, intrauterine growth restriction, and preeclampsia) or overzealous (consequences: placenta accreta, increta, and percreta) placentation. NK cells dominate in first trimester decidua and probably control extravillous cytotrophoblast (EVT) invasion. We examined this interaction in a novel way, using NK cells and villous explants from concordant first trimester pregnancies cocultured using a new collagen (two-dimensional) model of placentation. Decidual NK (dNK) cells exerted contact-independent inhibition of normal cytotrophoblast migration, associated with changes in the cytotrophoblast expression of metalloproteases-2 and -9, and plasminogen activator inhibitor-1. dNK cells did not affect EVT proliferation and apoptosis, and cell column formation. dNK cell effects were partially reversed by neutralizing Abs against IFN-gamma. We provide ex vivo human evidence of a direct role for dNK in modulating EVT differentiation as they form columns and then migrate from anchoring villi.


Assuntos
Movimento Celular , Interferon gama/fisiologia , Células Matadoras Naturais/fisiologia , Placenta/citologia , Primeiro Trimestre da Gravidez , Trofoblastos/citologia , Adulto , Comunicação Celular , Células Cultivadas , Técnicas de Cocultura , Feminino , Humanos , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , Placentação , Gravidez
8.
J Clin Endocrinol Metab ; 91(12): 4825-35, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17018655

RESUMO

CONTEXT: Gonadal steroids are key regulators of the extracellular matrix remodeling events that occur in the human endometrium during each menstrual cycle. The spatiotemporal expression of A Disintegrin And Metalloproteinase with ThromboSpondin repeats (ADAMTS)-1 in human endometrial stroma in vivo suggests that this novel metalloproteinase may contribute to this tightly regulated developmental process. OBJECTIVE: The objective of the study was to determine whether progesterone (P4), 17beta-estradiol (E2), or the nonaromatizable androgen dihydrotestosterone (DHT), alone or in combination, is capable of regulating ADAMTS-1 mRNA and protein levels in human endometrial stromal cells in a concentration- and time-dependent manner. DESIGN: A real-time quantitative PCR strategy and Western blotting were used to examine ADAMTS-1 mRNA and protein expression levels in primary cultures of human endometrial stromal cells. RESULTS: P4 and DHT but not E2 increased the levels of the ADAMTS-1 mRNA transcript and protein species (110 kDa) present in endometrial stromal cells in vitro in a concentration- and time-dependent manner. A combination of P4 and DHT resulted in an additional increase in stromal ADAMTS-1 expression, whereas E2 attenuated the regulatory effects of P4 and DHT in a concentration-dependent manner. The antisteroidal compounds, mifepristone (RU486) and hydroxyflutamide, were also found to inhibit specifically the P4- and DHT-mediated increase in ADAMTS-1 mRNA and protein expression levels in these primary cell cultures in a concentration-dependent manner, respectively. CONCLUSIONS: These studies demonstrate that progestins, androgens, and estrogens, alone and in combination, have distinct regulatory effects on ADAMTS-1 mRNA and protein expression levels in human endometrial stromal cells in vitro.


Assuntos
Proteínas ADAM/metabolismo , Androgênios/fisiologia , Endométrio/efeitos dos fármacos , Estrogênios/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hormônios Esteroides Gonadais/farmacologia , Progestinas/fisiologia , Células Estromais/efeitos dos fármacos , Proteína ADAMTS1 , Adulto , Antagonistas de Androgênios/farmacologia , Células Cultivadas , Di-Hidrotestosterona/farmacologia , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Endométrio/citologia , Endométrio/metabolismo , Estradiol/farmacologia , Feminino , Humanos , Pessoa de Meia-Idade , Progestinas/antagonistas & inibidores , Células Estromais/metabolismo , Fatores de Tempo
9.
Hum Reprod ; 21(8): 1990-9, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16675485

RESUMO

BACKGROUND: The pro-inflammatory cytokine, interleukin-1 beta (IL-1beta) promotes the proteolytic degradation of the extracellular matrix (ECM) of maternal decidua, a critical step in pregnancy that is counterbalanced by the expression of the anti-inflammatory cytokine, transforming growth factor-beta 1 (TGF-beta1). Recently, the inflammation-associated protein, ADAMTS-1, a member of the ADAMTS (A Disintegrin And Metalloproteinase with ThromboSpondin repeats) gene family of metalloproteinases has been assigned a central role in the formation and organization of tissues. In view of these observations, we have hypothesized that ADAMTS-1 contributes to the cytokine-mediated remodelling of decidual ECM. METHODS: The spatiotemporal expression of ADAMTS-1 in human endometrium was examined by immunohistochemistry. A quantitative-competitive (QC)-PCR strategy and western blot analysis was then employed to determine whether IL-1beta and TGF-beta1 regulate ADAMTS-1 mRNA and protein expression levels in primary cultures of stromal cells isolated from first trimester decidua. RESULTS: ADAMTS-1 expression is associated with decidualization of the endometrial stroma in vivo. IL-1beta increased whereas TGF-beta1 decreased ADAMTS-1 mRNA and protein levels in decidual stromal cell cultures in a concentration- and time-dependent manner. These regulatory effects were attenuated by function-perturbing antibodies specific for either cytokine. CONCLUSION: IL-1beta and TGF-beta1 differentially regulate ADAMTS-1 expression in human decidual stromal cells.


Assuntos
Proteínas ADAM/biossíntese , Decídua/metabolismo , Interleucina-1/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Proteína ADAMTS1 , Células Cultivadas , Decídua/citologia , Feminino , Humanos , Imuno-Histoquímica , Interleucina-1/imunologia , Ciclo Menstrual/fisiologia , Gravidez , RNA Mensageiro/metabolismo , Células Estromais/metabolismo , Fator de Crescimento Transformador beta/imunologia , Fator de Crescimento Transformador beta1
10.
J Clin Endocrinol Metab ; 89(3): 1459-66, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15001648

RESUMO

There is increasing evidence to suggest that the classical form of GnRH (GnRH I) and the second mammalian form of this hormone, GnRH II, play regulatory roles in human implantation and placentation. To date, the cellular distribution of these two hormones at the maternal-fetal interface remains poorly characterized. In these studies, we localized GnRH I and GnRH II expression in human placenta and decidua to distinct subpopulations of cells isolated from these tissues and in a chorionic villous/decidual tissue coculture system that mimics many of the cellular events of early placentation. GnRH I and GnRH II mRNA transcripts were detected in first-trimester placenta, whereas only GnRH I was detected in tissues obtained at term. Both hormones were further immunolocalized to the mononucleate villous and distinct subpopulations of extravillous cytotrophoblasts of the placenta in vivo and in vitro. In contrast, GnRH I but not GnRH II was expressed in the outer multinucleated syncytial trophoblast layer of first trimester chorionic villi and in cultures of villous cytotrophoblasts allowed to undergo differentiation and fusion in vitro. GnRH I and GnRH II were also found to be coexpressed in first-trimester decidua and primary cultures of decidual stromal cells. Collectively, these observations demonstrate that GnRH I and GnRH II have both common and discrete cellular distributions in the placenta and decidua and suggest that these two hormones are capable of eliciting their biological actions in an autocrine and/or paracrine manner within and between these maternal and fetal cellular compartments.


Assuntos
Decídua/fisiologia , Hormônio Liberador de Gonadotropina/genética , Células Cultivadas , Vilosidades Coriônicas/fisiologia , Técnicas de Cocultura , Decídua/citologia , Implantação do Embrião/fisiologia , Feminino , Expressão Gênica/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Gravidez , Primeiro Trimestre da Gravidez , RNA Mensageiro/análise , Células Estromais/citologia , Células Estromais/fisiologia , Trofoblastos/citologia , Trofoblastos/fisiologia
11.
J Clin Endocrinol Metab ; 88(10): 4781-90, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14557455

RESUMO

An intricate balance between the production of matrix metalloproteinases (MMPs) and their endogenous inhibitors, tissue inhibitors of metalloproteinases (TIMPs), modulates the overall proteolytic activity of trophoblasts during human implantation. In these studies we have examined the ability of classical GnRH I and the second form of this hormone (GnRH II) to regulate MMP-2, MMP-9, and TIMP-1 mRNA and protein levels in extravillous cytotrophoblasts propagated from explants of first trimester chorionic villi. GnRH I and GnRH II were found to increase MMP-2 and MMP-9 mRNA and protein levels in these primary cell cultures in a dose- and time-dependent manner using quantitative competitive-PCR and ELISA. In contrast, these two hormones decreased trophoblastic TIMP-1 mRNA and protein levels. Cetrorelix, a GnRH receptor antagonist, inhibited the regulatory effects of GnRH I, but not GnRH II, on MMP-2, MMP-9, and TIMP-1 expression in these cells. Collectively, these observations suggest that GnRH I and GnRH II differentially regulate MMP-2, MMP-9, and TIMP-1 expression in human trophoblasts, possibly via distinct receptor-mediated intracellular signaling pathways.


Assuntos
Hormônio Liberador de Gonadotropina/farmacologia , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , Inibidor Tecidual de Metaloproteinase-1/genética , Trofoblastos/efeitos dos fármacos , Trofoblastos/enzimologia , Células Cultivadas , Relação Dose-Resposta a Droga , Feminino , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/análogos & derivados , Antagonistas de Hormônios/farmacologia , Humanos , Metaloproteinase 9 da Matriz/metabolismo , Gravidez , Primeiro Trimestre da Gravidez , RNA Mensageiro/análise , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Trofoblastos/citologia
12.
J Clin Endocrinol Metab ; 88(8): 3806-15, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12915673

RESUMO

To date, the factors capable of regulating the coordinate expression of the urokinase-type plasminogen activator (uPA) and its endogenous inhibitor, plasminogen activator inhibitor (PAI-1), at the maternal-fetal interface remain poorly characterized. In these studies we examined the ability of the classical form of gonadotropin-releasing hormone (GnRH) I and the second, mammalian form of this hormone, GnRH II, to regulate uPA and PAI-1 mRNA and protein expression levels in cultures of stromal cells isolated from first trimester decidual tissues using quantitative competitive-PCR and ELISA, respectively. GnRH I and GnRH II increased uPA mRNA and protein expression levels in these primary cell cultures in a dose- and time-dependent manner. In contrast, GnRH I increased, whereas GnRH II decreased PAI-1 mRNA and protein expression levels in these cells. Cetrorelix, a GnRH receptor antagonist, inhibited the regulatory effects of GnRH I, but not GnRH II, on uPA and PAI-1 expression levels in these decidual stromal cell cultures. Taken together, these observations suggest that GnRH I and GnRH II differentially regulate the balance between uPA and PAI-1 expression levels in the human decidua, possibly via distinct receptor-mediated signaling pathways.


Assuntos
Decídua/fisiologia , Inativadores de Plasminogênio/fisiologia , Células Estromais/fisiologia , Ativador de Plasminogênio Tipo Uroquinase/fisiologia , Adulto , Separação Celular , Células Cultivadas , Primers do DNA , Decídua/citologia , Decídua/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Feminino , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/farmacologia , Antagonistas de Hormônios/farmacologia , Humanos , Gravidez , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/efeitos dos fármacos , Fatores de Tempo
13.
Biol Reprod ; 69(5): 1506-14, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12855604

RESUMO

Extensive remodeling of the extracellular matrix occurs in the ovary during the periovulatory period. Matrix metalloproteinases and their endogenous inhibitors, tissue inhibitors of metalloproteinases, are believed to play integral roles in this highly regulated series of cellular events, but their specific roles remain unclear. Recent cloning studies have identified a novel family of metalloproteinases, the ADAMTS (A Disintegrin And Metalloproteinase with ThromboSpondin motifs) family. The regulated expression of distinct ADAMTS subtypes has been shown to be required for tissue morphogenesis during embryonic development and for maintaining the integrity of tissues in the adult. In the present studies, we have determined that multiple ADAMTS subtypes are present in the bovine ovary using a reverse transcription-polymerase chain reaction strategy. In particular, ADAMTS-1, -2, -3, -4, -5 (also known as ADAMTS-11), -7, -8, and -9, but not ADAMTS-6, -10, or -12, mRNA transcripts were detected in granulosa cells of nonatretic ovarian follicles and corpora lutea. The levels of mRNA for these ovarian ADAMTS were up- or down-regulated or remained unchanged in the granulosa and/or theca cells of the dominant follicle following the preovulatory surge of gonadotropins, depending on the subtype and/or the cell compartment, and in the corpus luteum during the luteal phase of the estrous cycle. The complex expression patterns observed for the distinct ADAMTS subtypes in the granulosa and theca cells of the periovulatory follicle and in the luteal tissues of the bovine ovary suggest that these novel proteases mediate, at least in part, the remodeling events underlying folliculogenesis and ovulation and the formation, maintenance, and regression of the corpus luteum.


Assuntos
Corpo Lúteo/crescimento & desenvolvimento , Gonadotropinas/fisiologia , Luteólise/fisiologia , Metaloproteases/biossíntese , Folículo Ovariano/fisiologia , Ovulação/fisiologia , RNA Mensageiro/biossíntese , Animais , Southern Blotting , Bovinos , Primers do DNA , DNA Complementar/biossíntese , DNA Complementar/genética , Embrião de Mamíferos/fisiologia , Feminino , Fertilização in vitro , Células da Granulosa/metabolismo , Metaloproteases/genética , Gravidez , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tecais/metabolismo
14.
Int J Cancer ; 106(2): 172-7, 2003 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-12800191

RESUMO

The roles of the cadherins in the progression of ovarian cancer to the late stages of the disease state when malignant cells have disseminated within the peritoneal cavity remain poorly understood. In view of these observations, we have undertaken a comprehensive survey of the cadherin subtypes present in normal ovarian surface epithelium and peritoneum and in the tumors and peritoneal effusions of women diagnosed with Stage I or Stage II primary ovarian cancer using a degenerate cloning strategy for sequences highly conserved among this family of cell adhesion molecules. On the basis of the nucleotide sequences of the resultant PCR products, multiple cadherin subtypes (E-, N-, P-cadherin, and cadherin-4, -6, and -11) were found to be present in these normal and malignant tissues and cells. P-cadherin was determined to be the predominant cadherin subtype in normal peritoneum, peritoneal effusions and Stage II tumor masses. An increase in P-cadherin mRNA and protein expression levels in ovarian tumor masses with progression to later stages of the disease state was confirmed by Northern and Western blot analysis, respectively. In addition, we have determined that the cadherin-associated protein, known as beta-catenin, is expressed in normal peritoneum, ovarian tumors and malignant cell effusions obtained from women with Stage I or Stage II cancer. Immunoprecipitation studies demonstrated that P-cadherin was capable of interacting with beta-catenin in these normal and malignant tissues and cells. Collectively, these findings suggest that the regulated expression of P-cadherin/beta-catenin complexes in ovarian tumor cells may represent a key step in disease progression.


Assuntos
Caderinas/classificação , Caderinas/metabolismo , Neoplasias Ovarianas/metabolismo , Líquido Ascítico/metabolismo , Northern Blotting , Western Blotting , Caderinas/genética , Proteínas do Citoesqueleto/metabolismo , Primers do DNA/química , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Estadiamento de Neoplasias , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Peritônio/metabolismo , Derrame Pleural Maligno/metabolismo , Reação em Cadeia da Polimerase , Testes de Precipitina , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transativadores/metabolismo , beta Catenina
15.
Dev Biol ; 257(1): 41-54, 2003 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-12710956

RESUMO

E-cadherin and cadherin-11 are two members of the cadherin gene family of cell adhesion molecules that are differentially expressed during the aggregation, differentiation, and fusion of trophoblasts isolated from the human term placenta. E-cadherin expression is highest in cytotrophoblasts and decreases as these mononucleate cells undergo terminal differentiation and fusion. In contrast, cadherin-11 expression increases during the formation of multinucleated syncytium in these primary cultures. To define the role(s) of cadherin-11 in this developmental process, we examined the effects of ectopic cadherin-11 expression on the differentiation and fusion of JEG-3 choriocarcinoma cells, a mononucleate trophoblastic cell line. Cadherin-11 expression, but not the ectopic expression of the related cadherin subtype, cadherin-6, resulted in the formation of multinucleated syncytium in the transfected JEG-3 cell cultures. Multinucleated syncytium formation in the JEG-3 cells transfected with cadherin-11 was associated with a reduction in E-cadherin, alpha-, beta-, gamma-catenin, and p120(ctn) expression. Cadherin-11 also reduced cell proliferation and increased the levels of the mRNA transcript encoding the beta subunit of human chorionic gonadotropin, a biochemical marker of trophoblast differentiation, in these cultures. Furthermore, primary cytotrophoblasts cultured in the presence of antisense oligonucleotides specific for cadherin-11 maintained E-cadherin expression and did not undergo terminal differentiation and fusion with time in culture. Collectively, these observations demonstrate that cadherin-11 contributes to the morphological and functional differentiation of cultured mononucleate trophoblastic cells in a highly specific manner.


Assuntos
Caderinas/metabolismo , Diferenciação Celular/fisiologia , Trofoblastos/metabolismo , Fusão Celular , Linhagem Celular , DNA Antissenso/metabolismo , Feminino , Imunofluorescência , Humanos , Células Lúteas , Placenta/metabolismo , Gravidez , RNA Mensageiro/metabolismo , Trofoblastos/citologia
16.
J Clin Endocrinol Metab ; 88(2): 680-8, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12574199

RESUMO

Matrix metalloproteinases (MMPs) and their endogenous inhibitors, tissue-specific inhibitor of matrix metalloproteinases (TIMPs), play key roles in the cyclic remodeling events that occur in the human endometrium in preparation for pregnancy. To date, the factors capable of regulating the expression of MMPs and TIMPs in the human decidua remain poorly characterized. The spatiotemporal expression of GnRH in the human endometrium during the menstrual cycle and early pregnancy suggests that this hormone may have a regulatory role in the development of this dynamic tissue. In view of these observations, we have examined the ability of GnRH to regulate MMP-2, MMP-9, and TIMP-1 mRNA levels in primary cultures of human decidual stromal cells using a quantitative competitive PCR strategy. GnRH was capable of increasing MMP-2 and MMP-9 mRNA levels in these primary cell cultures in a dose-dependent manner. The GnRH antagonist, antide, was capable of inhibiting the GnRH-mediated increase in the levels of the MMP-2 and MMP-9 mRNA transcripts present in these decidual stromal cells in a dose-dependent manner. In contrast, GnRH or antide did not have a significant effect on TIMP-1 mRNA level in these primary cell cultures at any of the concentrations used in these studies. Taken together, these observations suggest that GnRH plays an integral role in human implantation, by virtue of its ability to regulate the balance between MMP and TIMP expression in decidual cells.


Assuntos
Decídua/citologia , Fármacos para a Fertilidade Feminina/farmacologia , Hormônio Liberador de Gonadotropina/farmacologia , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , Inibidor Tecidual de Metaloproteinase-1/genética , Células Cultivadas , Relação Dose-Resposta a Droga , Feminino , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Antagonistas de Hormônios/farmacologia , Humanos , Técnicas In Vitro , Oligopeptídeos/farmacologia , Gravidez , RNA Mensageiro/análise , Células Estromais/citologia , Células Estromais/fisiologia
17.
J Clin Endocrinol Metab ; 87(12): 5594-603, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12466358

RESUMO

The regulated expression of the urokinase-type plasminogen activator (uPA) and plasminogen activator inhibitor (PAI-1) is believed to modulate the invasive capacity of human trophoblastic cells in vitro and in vivo. To date, the factors capable of regulating the expression of uPA and PAI-1 in these cells remain poorly characterized. In these studies, we have examined the ability of the classical mammalian GnRH I and the second form of GnRH (GnRH II) to regulate uPA and PAI-1 mRNA and protein expression levels in primary cultures of human extravillous cytotrophoblasts using quantitative competitive PCR and ELISA, respectively. Both GnRH I and II increased uPA and concomitantly decreased PAI-1 mRNA and protein expression levels in our extravillous cytotrophoblast cultures in a dose- and time-dependent manner. Cetrorelix, a peptide GnRH antagonist specific for the GnRH I receptor, was capable of inhibiting the regulatory effects of GnRH I, but not GnRH II on uPA and PAI-1 expression levels in primary cell cultures. Taken together, these observations suggest that GnRH I and GnRH II may facilitate trophoblast invasion by increasing the ratio of uPA/PAI-1 expression via interactions with two distinct GnRH receptors.


Assuntos
Hormônio Liberador de Gonadotropina/farmacologia , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Trofoblastos/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Células Cultivadas , Meios de Cultura/metabolismo , Hormônio Liberador de Gonadotropina/análogos & derivados , Antagonistas de Hormônios/farmacologia , Humanos , Inibidor 1 de Ativador de Plasminogênio/genética , Isoformas de Proteínas/farmacologia , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/metabolismo , Fatores de Tempo , Ativador de Plasminogênio Tipo Uroquinase/genética
18.
Biol Reprod ; 67(5): 1572-9, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12390890

RESUMO

Herpes simplex virus (HSV)-1 has been discovered in placental tissue from spontaneous miscarriages, but reports of transplacental transmission and fetal infection are extremely rare. Previously, we demonstrated that the villous syncytiotrophoblast, which forms a continuous layer between the maternal and fetal circulation, is resistant to HSV entry. Here, we tested our hypothesis that the villous syncytiotrophoblast prevents transplacental transmission of HSV secondary to decreased expression of HSV entry mediators (HveA, HveB, and HveC). In addition, we investigated the ability of HSV to infect extravillous trophoblast cells, which mediate placental attachment to the uterine wall, and the expression of HSV receptors in these cells. We performed fluorescence-activated cell sorting (FACS) analyses and immunostaining to demonstrate that HveA, HveB, and HveC were not expressed in third-trimester villous trophoblast cells. Consequently, villous explants obtained from third-trimester placentas were resistant to infection by a recombinant HSV-1 vector, HSV-1 KOS, but approximately 20% of mesenchymal cells within the villous core were infected when villous explants were pretreated with trypsin to disrupt the villous trophoblast layer. Conversely, FACS analysis and immunostaining demonstrated that extravillous trophoblast cells expressed HveA, HveB, and HveC, and these cells were efficiently infected by HSV vectors. Infection of extravillous trophoblast cells by HSV-1 was not reduced when the cells were pretreated with an antibody against HveA but was partially reduced when the cells were pretreated with antibodies directed against HveB and HveC. Thus, the decreased expression of herpesvirus entry mediators in villous syncytiotrophoblast prevents placental villous infection, thereby limiting maternal-fetal transmission of HSV.


Assuntos
Herpes Simples/transmissão , Transmissão Vertical de Doenças Infecciosas , Trofoblastos/fisiologia , Trofoblastos/virologia , Separação Celular , Células Cultivadas , Vilosidades Coriônicas , Feminino , Citometria de Fluxo , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/crescimento & desenvolvimento , Humanos , Placenta/citologia , Gravidez , Primeiro Trimestre da Gravidez , Terceiro Trimestre da Gravidez , Receptores do Fator de Necrose Tumoral/metabolismo , Membro 14 de Receptores do Fator de Necrose Tumoral , Receptores Virais/metabolismo , Trofoblastos/citologia , Trofoblastos/efeitos dos fármacos , Tripsina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...