Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
FASEB J ; 37(10): e23204, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37738042

RESUMO

Placental insufficiency disorders, including preeclampsia and intrauterine growth restriction, are major obstetric complications that can have devastating effects on both the mother and the fetus. These syndromes have underlying poor placental trophoblast cell invasion into uterine tissues. Placental invasion is controlled by many hormones and growth factors. Myostatin (MSTN) is a transforming growth factor-ß superfamily member recognized for its important role in muscle growth control. MSTN has also been shown to be secreted and functioning in the placenta, and its serum and/or placental levels were found to be upregulated in preeclampsia and intrauterine growth restriction. Considering that the mechanistic role of MSTN in placentation remains poorly understood, we hypothesized that MSTN uses ALK4/5-SMAD2/3/4 signaling to increase human trophoblast invasion through a group of epithelial-mesenchymal transition genes including SERPINE2, PAI-1, and SOX4. mRNA sequencing of control and MSTN-treated primary human trophoblast cells (n = 5) yielded a total of 610 differentially expressed genes (false discovery rate <0.05) of which 380 genes were upregulated and 230 were downregulated. These differentially expressed genes were highly enriched in epithelial-mesenchymal transition genes, and a subset including SERPINE2, PAI-1, and SOX4 was investigated for its role in MSTN-induced trophoblast cell invasion. We found that MSTN induced upregulation of SERPINE2 via ALK4/5-SMAD2/3/4 signaling; however, SMAD2 was not involved in MSTN-induced PAI-1 upregulation. SOX4 was involved in MSTN-induced upregulation of SERPINE2, but not PAI-1. Collectively, this study discovers novel molecular mechanisms of MSTN-induced human trophoblast cell invasion and provides insight into the functional consequences of its dysregulation in placental insufficiency disorders.


Assuntos
Miostatina , Insuficiência Placentária , Pré-Eclâmpsia , Feminino , Humanos , Gravidez , Transição Epitelial-Mesenquimal , Retardo do Crescimento Fetal , Peptídeos e Proteínas de Sinalização Intercelular , Miostatina/genética , Placenta , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidores de Serina Proteinase , Serpina E2/genética , Fatores de Transcrição SOXC , Trofoblastos
2.
Biol Reprod ; 106(6): 1267-1277, 2022 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-35020826

RESUMO

Placental insufficiency disorders are major obstetric complications that share a common phenomenon of poor placental trophoblast cell invasion and remodeling of uterine tissues. Myostatin is a transforming growth factor (TGF)-ß superfamily member well known for its important role in muscle growth control. Myostatin is also produced in the placenta and has been shown to regulate some trophoblast functions. However, its roles in placental development are still poorly understood. In this study, we tested the hypothesis that myostatin increases trophoblast cell invasion by upregulating N-cadherin via SMAD2/3-SMAD4 signaling. Primary and immortalized (HTR8/SVneo) trophoblast cells were used as study models. Matrigel-coated transwell invasion assays were used to study the effects of recombinant human myostatin on trophoblast cell invasion. Reverse transcription quantitative real-time polymerase chain reaction and Western blot were used to measure myostatin effects on N-cadherin mRNA and protein levels, respectively. Small inhibitor molecules as well as siRNA-mediated knockdown were used to block myostatin receptor and downstream signaling, respectively. Data were analyzed either by unpaired Student T test or one-way analysis of variance followed by Newman Keuls test for multiple group comparisons. Myostatin significantly increased primary and HTR8/SVneo trophoblast cell invasion. Moreover, myostatin upregulated N-cadherin mRNA and protein levels in a time-dependent manner in both study models. These effects were blocked by inhibition of TGF-ß type I receptors as well as siRNA-mediated knockdown of SMAD2/3 combined or common SMAD4. Importantly, myostatin-induced trophoblast cell invasion was abolished by knockdown of N-cadherin, SMAD2/3, or SMAD4. Myostatin may increase human trophoblast cell invasion by upregulating N-cadherin via SMAD2/3-SMAD4 signaling.


Assuntos
Caderinas , Trofoblastos , Caderinas/genética , Caderinas/metabolismo , Movimento Celular , Feminino , Humanos , Miostatina/metabolismo , Placenta/metabolismo , Gravidez , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Proteína Smad4/metabolismo , Proteína Smad4/farmacologia , Fator de Crescimento Transformador beta/metabolismo , Trofoblastos/metabolismo
3.
Biol Reprod ; 107(2): 458-473, 2022 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-35403677

RESUMO

Hyaluronan is a structural component of the expanded cumulus matrix, and hyaluronan synthase 2 is the major enzyme for the synthesis of hyaluronan in humans. Versican cross-links the hyaluronan-rich matrix to cumulus cells and is critical for successful ovulation. Activin A is a critical intrafollicular regulator of ovarian function. Although activin A has been shown to promote cumulus matrix expansion in mice, the functional role of activin A in the regulation of cumulus expansion in the human ovary remains to be elucidated. Using primary and immortalized human granulosa-lutein cells as study models, we provide the first data showing that activin A increased the production of hyaluronan by upregulating the expression of hyaluronan synthase 2 in these cells. Additionally, activin A also promoted the expression of the hyaluronan-binding protein versican. Moreover, using inhibitor- and small interfering RNA-mediated inhibition approaches, we found that these stimulatory effects of activin A are most likely mediated through the type I receptor activin receptor-like kinase (ALK4)-mediated Sma- and Mad-related protein (SMAD2)/SMAD3-SMAD4 signaling pathway. Notably, the chromatin immunoprecipitation analyses demonstrated that SMAD4 could bind to human hyaluronan synthase 2 and VERSICAN promoters. The results obtained from this in vitro study suggest that locally produced activin A plays a functional role in the regulation of hyaluronan production and stabilization in human granulosa-lutein cells.


Assuntos
Ácido Hialurônico , Versicanas , Ativinas , Células Cultivadas , Feminino , Células da Granulosa/metabolismo , Humanos , Hialuronan Sintases/genética , Hialuronan Sintases/metabolismo , Ácido Hialurônico/metabolismo , Versicanas/genética , Versicanas/metabolismo , Versicanas/farmacologia
4.
FASEB J ; 35(12): e22028, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34739154

RESUMO

The interplay between growth factors, signaling pathways and transcription factors during placental development is key to controlling trophoblast differentiation. Bone morphogenetic protein 2 (BMP2) has been implicated in trophoblast invasion and spiral artery remodeling during early placental development. However, the molecular mechanisms by which these are accomplished have not been fully elucidated, particularly for transcriptional regulation of key transcription factors. Here, we identified SOX4 as a direct target gene induced by BMP2 in first-trimester placental trophoblasts. Analysis of single-cell RNA-seq data from first-trimester placentas and decidua tissues revealed that SOX4 expression is mainly localized in extravillous trophoblast and decidual stromal cells. Moreover, gain- and loss-of-function approaches demonstrated that SOX4 exerts a pro-invasive role in human trophoblasts, and this effect contributes to BMP2-enhanced trophoblast invasion. Importantly, we found that SOX4 was required for BMP2-induced regulation of a subset of genes associated with cell migration and extracellular matrix organization. We also show that SOX4-dependent regulation of the BMP2 target SERPINE2 occurs via binding of SOX4 to regulatory elements such as enhancers, thereby promoting BMP2-induced trophoblast invasion. In conclusion, these findings uncover a novel mechanism involving SOX4 that shapes the BMP2-regulated transcriptional network during invasive trophoblast development.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Diferenciação Celular , Regulação da Expressão Gênica no Desenvolvimento , Placenta/patologia , Placentação , Fatores de Transcrição SOXC/metabolismo , Serpina E2/metabolismo , Trofoblastos/patologia , Proteína Morfogenética Óssea 2/genética , Feminino , Humanos , Placenta/metabolismo , Gravidez , Fatores de Transcrição SOXC/genética , Serpina E2/genética , Trofoblastos/metabolismo
5.
FASEB J ; 34(2): 3151-3164, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31908038

RESUMO

Extravillous cytotrophoblasts (EVTs) invade into the uterine wall and remodel spiral arteries for proper placentation. Studies from us and others have demonstrated that the transforming growth factor-ß superfamily member bone morphogenetic protein 2 (BMP2) plays important roles in endometrial decidualization and trophoblast cell invasion. However, BMP2 has also been shown to regulate endothelial cell migration and capillary-like tube formation, as has its downstream signaling molecule inhibitor of DNA binding 1 (ID1). Interestingly, insulin-like growth factor binding protein 3 (IGFBP3) also promotes cell migration and angiogenesis in endothelial precursor cell. Moreover, Id1 has a regulatory effect on Igfbp3 expression in rat prostate epithelial cells. However, whether ID1 and IGFBP3 are integrated in BMP2 signaling and involved in the regulation of trophoblast invasive and endovascular differentiation remains unknown. The objective of our study was to examine the effects of BMP2 on ID1 and IGFBP3 expression and their roles in BMP2-regulated human trophoblast invasion and endothelial-like tube formation. Primary and immortalized (HTR8/SVneo) cultures of human trophoblast cells were employed as study models. BMP2 treatment increased ID1 and IGFBP3 mRNA and protein levels in HTR8/SVneo and primary human EVT cells. Intriguingly, ID1 was essential for BMP2-induced IGFBP3 upregulation in both study models, and BMP2-induced trophoblast invasion was attenuated by knockdown of either ID1 or IGFBP3. In addition, BMP2 significantly increased endothelial-like tube formation and knockdown of ID1 and IGFBP3 reduced basal and BMP2-induced tube formation in HTR8/SVneo cells. Similarly, BMP2 increased placenta growth factor (PlGF) production in HTR8/SVneo cells and these effects were attenuated by knockdown of ID1 or IGFBP3. Our results reveal that BMP2 promotes trophoblast cell invasion and endothelial-like tube formation by ID1-mediated IGFBP3 upregulation.


Assuntos
Proteína Morfogenética Óssea 2/farmacologia , Diferenciação Celular , Movimento Celular , Células Endoteliais/metabolismo , Trofoblastos/metabolismo , Linhagem Celular , Células Cultivadas , Células Endoteliais/citologia , Humanos , Proteína 1 Inibidora de Diferenciação/genética , Proteína 1 Inibidora de Diferenciação/metabolismo , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Fator de Crescimento Placentário/metabolismo , Trofoblastos/citologia , Trofoblastos/efeitos dos fármacos , Trofoblastos/fisiologia , Regulação para Cima
6.
FASEB J ; 34(7): 9419-9432, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32501590

RESUMO

Neuropeptide FF (NPFF) is well-known for its roles in the central nervous system. Despite studies demonstrating that NPFF receptor 2 (NPFFR2) mRNA is highest in placenta, nothing is known about NPFF-NPFFR2 functions in placental development. Here, we investigated the effects of NPFF-NPFFR2 on expression of syncytial [human chorionic gonadotropin (hCG) ß] and fusogenic [syncytin 1, syncytin 2, and glial cells missing 1 (GCM1)] genes in first trimester primary human cytotrophoblast cells. By analyzing two publicly available microarray data sets, we found that NPFF is consistently expressed throughout gestation whereas NPFFR2 increases in first trimester and is elevated in placenta samples from women with preeclampsia. Immunohistochemistry showed that NPFFR2, syncytin 1/2, and GCM1 each displayed unique patterns of expression among different trophoblast populations in first trimester placenta. Treatment of primary human cytotrophoblast cells with NPFF increased the mRNA and protein levels of hCG ß, syncytin 1, syncytin 2, and GCM1; and knockdown of NPFFR2 abolished these effects. Interestingly, GCM1 mediated NPFF-induced upregulation of syncytin 1 and syncytin 2, but not hCG ß, in primary human cytotrophoblasts. Our results demonstrate that NPFF acts via NPFFR2 to enhance production of hCG ß and promote GCM1-dependent expression of syncytin 1 and 2 in human cytotrophoblasts.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Produtos do Gene env/metabolismo , Oligopeptídeos/farmacologia , Pré-Eclâmpsia/patologia , Proteínas da Gravidez/metabolismo , Fatores de Transcrição/metabolismo , Trofoblastos/patologia , Biomarcadores/metabolismo , Proteínas de Ligação a DNA/genética , Feminino , Perfilação da Expressão Gênica , Produtos do Gene env/genética , Humanos , Antagonistas de Entorpecentes/farmacologia , Placentação , Pré-Eclâmpsia/tratamento farmacológico , Pré-Eclâmpsia/genética , Pré-Eclâmpsia/metabolismo , Gravidez , Proteínas da Gravidez/genética , Primeiro Trimestre da Gravidez , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/metabolismo , Fatores de Transcrição/genética , Trofoblastos/efeitos dos fármacos , Trofoblastos/metabolismo
7.
Exp Cell Res ; 382(2): 111471, 2019 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-31229504

RESUMO

The highly invasive and metastatic properties of ovarian cancer make it the leading cause of death among gynecological cancers. Elevated levels of activin A and its receptors have been found in ovarian tumors and are associated with reduced survival in ovarian cancer patients. The role of activin A in promoting ovarian cancer cell migration and invasion has been previously reported, however, the underlying molecular mechanisms remain largely unknown. Here, we show that treatment of SKOV3 and OVISE cells with activin A decreases the expression of E-cadherin. These effects are completely diminished by inhibition or knockdown of the activin type I receptor. Treatment with activin A activates SMAD2/3 signaling but does not alter MEK-ERK1/2 or PI3K/AKT signaling pathway activity. Knockdown of SMAD2, SMAD3 or SMAD4 abolishes the downregulation of E-cadherin by activin A. Moreover, activin A treatment induces the expression of transcription factors SNAIL and SLUG, which mediate the suppressive effects of activin A on E-cadherin expression. Importantly, forced-expression of E-cadherin inhibits both basal and activin A-induced cell migration. Taken together, our results suggest that activin A downregulates E-cadherin expression by upregulating SLUG and SNAIL expression via SMAD2/3-SMAD4-dependent signaling. Loss of E-cadherin contributes to activin A-induced ovarian cancer cell migration.


Assuntos
Ativinas/metabolismo , Antígenos CD/metabolismo , Caderinas/metabolismo , Movimento Celular , Neoplasias Ovarianas/patologia , Receptores de Ativinas Tipo I/metabolismo , Antígenos CD/genética , Caderinas/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Regulação para Baixo/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Ovarianas/genética , Fosforilação , Transdução de Sinais , Proteínas Smad/metabolismo , Fatores de Transcrição da Família Snail/metabolismo , Regulação para Cima/genética
8.
Proc Natl Acad Sci U S A ; 112(15): 4743-8, 2015 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-25825716

RESUMO

Polycystic ovary syndrome (PCOS) is one of the most common female endocrine disorders and a leading cause of female subfertility. The mechanism underlying the pathophysiology of PCOS remains to be illustrated. Here, we identify two alternative splice variants (ASVs) of the androgen receptor (AR), insertion and deletion isoforms, in granulosa cells (GCs) in ∼62% of patients with PCOS. AR ASVs are strongly associated with remarkable hyperandrogenism and abnormalities in folliculogenesis, and are absent from all control subjects without PCOS. Alternative splicing dramatically alters genome-wide AR recruitment and androgen-induced expression of genes related to androgen metabolism and folliculogenesis in human GCs. These findings establish alternative splicing of AR in GCs as the major pathogenic mechanism for hyperandrogenism and abnormal folliculogenesis in PCOS.


Assuntos
Processamento Alternativo , Mutação INDEL , Síndrome do Ovário Policístico/genética , Receptores Androgênicos/genética , Adulto , Sequência de Bases , Células Cultivadas , Desidroepiandrosterona/sangue , Feminino , Perfilação da Expressão Gênica , Estudo de Associação Genômica Ampla , Células da Granulosa/metabolismo , Células HEK293 , Humanos , Hiperandrogenismo/sangue , Hiperandrogenismo/genética , Oogênese/genética , Folículo Ovariano/fisiopatologia , Síndrome do Ovário Policístico/sangue , Síndrome do Ovário Policístico/fisiopatologia , Isoformas de Proteínas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Testosterona/sangue
9.
J Cell Physiol ; 231(1): 111-9, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26031779

RESUMO

Connexin43 (Cx43) has been shown to regulate cell proliferation and its downregulation is correlated with poor prognosis and survival in several types of human cancer. Cx43 expression levels are frequently downregulated in human ovarian cancer, suggesting a potential role for Cx43 in regulating the progression of this disease. Epidermal growth factor (EGF) is a well-characterized hormone that stimulates ovarian cancer cell proliferation. Although EGF is able to regulate Cx43 expression in other cell types, it is unclear whether EGF can regulate Cx43 expression in ovarian cancer cells. Additionally, it remains unknown whether Cx43 is involved in EGF-stimulated ovarian cancer cell proliferation. In the present study, we demonstrate that treatment with EGF upregulates Cx43 expression in two ovarian cancer cell lines, SKOV3 and OVCAR4. Although treatment with EGF activates both ERK1/2 and Akt signaling pathways, pharmacological inhibition and siRNA-mediated knockdown suggest that only the activation of Akt1 is required for EGF-induced Cx43 upregulation. Functionally, Cx43 knockdown enhanced basal and EGF-induced cell proliferation, whereas the proliferative effects of EGF were reduced by Cx43 overexpression. Co-treatment with the gap junction inhibitor carbenoxolone did not alter the suppressive effects of Cx43 overexpression on EGF-induced cell proliferation, suggesting a gap junction-independent mechanism. This study reveals an important role for Cx43 as a negative regulator of EGF-induced human ovarian cancer cell proliferation.


Assuntos
Proliferação de Células/efeitos dos fármacos , Conexina 43/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Junções Comunicantes/efeitos dos fármacos , Neoplasias Ovarianas/metabolismo , Comunicação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Junções Comunicantes/metabolismo , Humanos , Transdução de Sinais/efeitos dos fármacos
10.
Mol Hum Reprod ; 22(2): 119-29, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26660506

RESUMO

STUDY HYPOTHESIS: We hypothesized that Runt-related transcription factor 2 (RUNX2), matrix metalloproteinase (MMP)2 and MMP9 are involved in basal and gonadotrophin-releasing hormone (GnRH)-induced human extravillous trophoblast (EVT) cell invasion. STUDY FINDING: Our finding indicates that GnRH-induced RUNX2 expression enhances the invasive capacity of EVT cells by modulating the expression of MMP2 and MMP9. WHAT IS KNOWN ALREADY: GnRH is expressed in first-trimester placenta and exerts pro-invasive effects on EVT cells in vitro. RUNX2 regulates MMP2 and MMP9 expression and is often associated with invasive phenotypes. STUDY DESIGN, SAMPLES/MATERIALS, METHODS: First-trimester human placenta (n = 9) was obtained from women undergoing elective termination of pregnancy. The localization of RUNX2, MMP2 and MMP9 in first-trimester human placenta was examined by immunohistochemistry. Primary or immortalized (HTR-8/SVneo) EVT cells were treated alone or in combination with GnRH, GnRH antagonist Antide, MAPK kinase inhibitor PD98095, phosphatidylinositol 3-kinase inhibitor LY294002, MMP2/9 inhibitor or small interfering RNAs (siRNAs) targeting RUNX2, MMP2 and/or MMP9. Protein and mRNA levels were measured by western blot and RT-PCR, respectively. Cell invasiveness was evaluated by transwell Matrigel or collagen I invasion assays. MAIN RESULTS AND THE ROLE OF CHANCE: RUNX2, MMP2 and MMP9 were detected in the cell column regions of human first-trimester placental villi. GnRH treatment increased RUNX2 mRNA and protein levels in HTR-8/SVneo cells and primary EVTs, and these effects were attenuated by co-treatment with Antide, PD98095 or LY294002. Down-regulation of RUNX2 by siRNA reduced basal and GnRH-induced MMP2/9 expression and cell invasion. Moreover, pharmacological inhibition or siRNA-mediated knockdown of MMP2/9 reduced basal and GnRH-induced cell invasion. LIMITATIONS, REASONS FOR CAUTION: The lack of an in vivo model is the major limitation of our in vitro study. WIDER IMPLICATIONS OF THE FINDINGS: Our findings provide important insight into the functions of the GnRH - GnRH receptor system in early implantation and placentation. LARGE SCALE DATA: Not applicable. STUDY FUNDING AND COMPETING INTERESTS: This research was supported by Canadian Institutes of Health Research (Grant #143317) to P.C.K.L. The authors have nothing to disclose.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core/genética , Hormônio Liberador de Gonadotropina/farmacologia , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , RNA Mensageiro/genética , Trofoblastos/efeitos dos fármacos , Aborto Legal , Adulto , Linhagem Celular Transformada , Movimento Celular/efeitos dos fármacos , Vilosidades Coriônicas/metabolismo , Cromonas/farmacologia , Subunidade alfa 1 de Fator de Ligação ao Core/antagonistas & inibidores , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Decídua/citologia , Decídua/metabolismo , Feminino , Flavonoides/farmacologia , Regulação da Expressão Gênica , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Morfolinas/farmacologia , Oligopeptídeos/farmacologia , Gravidez , Primeiro Trimestre da Gravidez , Cultura Primária de Células , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Trofoblastos/citologia , Trofoblastos/metabolismo
11.
Reproduction ; 152(3): 205-13, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27335131

RESUMO

Lysyl oxidase (LOX), a key enzyme in the formation and stabilization of the extracellular matrix, is expressed in granulosa cells and plays a critical role in the regulation of granulosa cell differentiation, oocyte maturation and ovulation. To date, the regulation of LOX expression in human granulosa cells remains largely unknown. In this study, using primary and immortalized human granulosa lutein cells, we demonstrated that transforming growth factor (TGF)-ß1 (TGFB1) upregulated LOX expression and downregulated microRNA-29a (MIR29A) expression via a TGF-ß type I receptor-mediated signaling pathway. Additionally, we showed that MIR29A downregulated the expression of LOX in both types of cells. Furthermore, the downregulation of MIR29A contributed to the TGFB1-induced increase in LOX expression because the inhibition of MIR29A with a MIR29A inhibitor not only reversed the MIR29A-induced downregulation of LOX but also enhanced the TGFB1-induced upregulation of LOX. Our findings suggest that TGFB1 and MIR29A may play essential roles in the regulation of extracellular matrix remodeling during the periovulatory phase.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Células da Granulosa/metabolismo , Células Lúteas/metabolismo , MicroRNAs/antagonistas & inibidores , Proteína-Lisina 6-Oxidase/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Células Cultivadas , Regulação para Baixo , Feminino , Células da Granulosa/efeitos dos fármacos , Humanos , Células Lúteas/efeitos dos fármacos , MicroRNAs/genética
12.
Reproduction ; 152(4): 293-301, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27530347

RESUMO

Lysyl oxidase (LOX) is the key enzyme involved in the crosslinking of collagen and elastin that is essential for the formation of extracellular matrix (ECM). LOX-mediated ECM remodeling plays a critical role in follicle development, oocyte maturation and corpus luteum formation. To date, the regulation of LOX in human ovary has never been elucidated. Activin A and its functional receptors are highly expressed in ovarian follicles from an early developmental stage. They locally regulate follicle progression. The aim of this study was to investigate the effects of activin A on the expression of LOX and its extracellular enzyme activity in primary and immortalized human granulosa-lutein cells obtained from patients undergoing an in vitro fertilization procedure. We demonstrated that activin A significantly upregulated the expression of connective tissue growth factor (CTGF) and LOX via an activin/TGF-ß type I receptor mediated-signaling pathway. Using a target depletion small interfering RNA knockdown approach, we further confirmed that the upregulation of CTGF expression resulted in an activin-A-induced increases in LOX expression and activity. These findings may provide insight into the mechanisms by which intrafollicular growth factors regulate the expression of LOX for ECM formation and tissue remodeling in the human ovary.


Assuntos
Ativinas/farmacologia , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Células da Granulosa/enzimologia , Células Lúteas/enzimologia , Folículo Ovariano/enzimologia , Proteína-Lisina 6-Oxidase/metabolismo , Células Cultivadas , Fator de Crescimento do Tecido Conjuntivo/genética , Feminino , Células da Granulosa/efeitos dos fármacos , Humanos , Células Lúteas/efeitos dos fármacos , Folículo Ovariano/efeitos dos fármacos , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
13.
Biochem Biophys Res Commun ; 461(1): 128-35, 2015 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-25869072

RESUMO

Transforming growth factor-α (TGF-α), like epidermal growth factor (EGF) and amphiregulin (AREG) binds exclusively to EGF receptor (EGFR). We have previously demonstrated that EGF, AREG and TGF-α down-regulate E-cadherin and induce ovarian cancer cell invasion, though whether these ligands use the same molecular mediators remains unknown. We now show that, like EGF, TGF-α- and AREG-induced E-cadherin down-regulation involves both EGFR and HER2. However, in contrast to EGF and AREG, the transcription factor Snail is not required for TGF-α-induced E-cadherin down-regulation. This study shows that TGF-α uses common and divergent molecular mediators to regulate E-cadherin expression and cell invasion.


Assuntos
Caderinas/metabolismo , Família de Proteínas EGF/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Anfirregulina , Linhagem Celular Tumoral , Regulação para Baixo , Feminino , Humanos , Invasividade Neoplásica , Receptor ErbB-2/metabolismo , Transdução de Sinais , Fatores de Transcrição da Família Snail
14.
F S Sci ; 4(1): 47-55, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36152991

RESUMO

OBJECTIVE: To study the role of interleukin (IL)-1ß and the plasminogen activating (PA) system members in endometriotic stromal cell (ESC) migration/invasion. DESIGN: Primary cultures of ESCs. SETTING: Tertiary referral center for endometriosis and pelvic pain. PATIENT(S): Patients with surgically excised endometriosis. INTERVENTION(S): Interleukin-1ß stimulation of primary cultures of ESCs and knockdown of the PA system members urokinase plasminogen activator (uPA), uPA receptor, and plasminogen activator inhibitor-1 (PAI-1). MAIN OUTCOME MEASURE(S): Invasion/migration assays. RESULT(S): In primary cultures, IL-1ß-stimulated ESC production of the PA system members uPA, uPA receptor, and PAI-1. Interleukin-1ß also enhanced ESC migration and invasion, and these effects were inhibited by the IL-1 receptor-1 antagonist anakinra. Knockdown of each of the 3 PA system members also inhibited ESC migration and invasion. Knockdown of these PA system members further attenuated the impact of IL-1ß on migration and invasion, suggesting that they mediated the promigration and proinvasion effects of IL-1ß. To supplement the cell culture work, immunohistochemistry was performed on tissue sections of endometriotic epithelium/stroma: uPA, PAI-1, and IL-1ß histoscores were not found to be correlated with each other. CONCLUSION(S): In primary cultures of ESCs, IL-1ß induces migration and invasion, which is mediated by PA system members and inhibited by the drug anakinra. However, the immunohistochemistry expression of IL-1ß, urokinase plasminogen inhibitor-1, and PAI-1 were not correlated, suggesting other regulatory mechanisms for PA system members. Inhibition of IL-1ß (e.g., with anakinra) may have potential as a novel treatment approach for the migration/invasion of endometriosis.


Assuntos
Endometriose , Inibidor 1 de Ativador de Plasminogênio , Feminino , Humanos , Interleucina-1beta/farmacologia , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/genética , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Plasminogênio , Endometriose/genética , Proteína Antagonista do Receptor de Interleucina 1 , Movimento Celular
15.
Front Cell Dev Biol ; 10: 968166, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36092732

RESUMO

Vascular endothelial-cadherin (VE-cadherin) is an essential component that regulates angiogenesis during corpus luteum formation. Amphiregulin (AREG) and transforming growth factor ß1 (TGF-ß1) are two intrafollicular factors that possess opposite functions in directing corpus luteum development and progesterone synthesis in human granulosa-lutein (hGL) cells. However, whether AREG or TGF-ß1 regulates the VE-cadherin expression and subsequent angiogenesis in the human corpus luteum remains to be elucidated. Results showed that hGL cells cultured on Matrigel spontaneously formed capillary-like and sprout-like microvascular networks. Results of specific inhibitor treatment and small interfering RNA-mediated knockdown revealed that AREG promoteed microvascular-like formation in hGL cells by upregulating the VE-cadherin expression mediated by the epidermal growth factor receptor (EGFR)-extracellular signal-regulated kinase1/2 (ERK1/2) signaling pathway. However, TGF-ß1 suppressed microvascular-like formation in hGL cells by downregulating VE-cadherin expression mediated by the activin receptor-like kinase (ALK)5-Sma- and Mad-related protein (SMAD)2/3/4 signaling pathway. Collectively, this study provides important insights into the underlying molecular mechanisms by which TGF-ß1 and AREG differentially regulate corpus luteum formation in human ovaries.

16.
BMC Physiol ; 11: 18, 2011 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-22168796

RESUMO

This report presents evidence for the specificities of select commercially available HOXA4 antibodies in regards to concerns about the specificity of the HOXA4 antibody used by Lillvis et al. (Regional expression of HOXA4 along the aorta and its potential role in human abdominal aortic aneurysms. BMC Physiol 2011, 11:9). Using an antibody characterized extensively by us, Lillvis et al. report detecting HOXA4 at a size of 33 kDa despite our previous reports that HOXA4 is detected at ~37-39 kDa and that the ~30-33 kDa band is non-specific. Using small interfering RNA targeting HOXA4, forced expression of full-length HOXA4 and HOXA4-positive and -negative ovarian cancer cell lines, we confirm our previous findings that the ~30-33 kDa band is non-specific and that HOXA4 is detected at ~37-39 kDa. Moreover, we demonstrate that HOXA4 small interfering RNA reduces the ~37-39 kDa HOXA4 band, but not the ~30-33 kDa non-specific band, in a human acute monocytic leukemia cell line used by Lillvis et al. Western blot analysis performed with two additional commercially available HOXA4 antibodies also detected HOXA4 at ~37-39 kDa. Lastly, immunofluorescent staining of a HOXA4-negative ovarian cancer cell line with the antibody used by Lillvis et al. yields strong perinuclear staining, similar to that observed by Lillvis et al., which cannot be attributed to HOXA4. Our results highlight and briefly discuss the importance of careful antibody validation and selection for use in various applications.


Assuntos
Aorta/metabolismo , Aneurisma da Aorta Abdominal/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genes Homeobox , Proteínas de Homeodomínio/biossíntese , Animais , Feminino , Humanos , Masculino
17.
Front Cell Dev Biol ; 9: 768669, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34970543

RESUMO

Many pregnancy disorders, including early-onset preeclampsia (EOPE), are associated with defects in placental trophoblast cell invasion and differentiation during early placental development. Bone morphogenetic protein 2 (BMP2) belongs to the TGF-ß superfamily and controls various physiological and developmental processes. However, the expression of BMP2 in the placenta and underlying molecular mechanisms of how BMP2 regulates trophoblast function remain unclear. In this study, we analyzed several publicly available microarray and RNA-seq datasets and revealed differences in expression of TGF-ß superfamily members between gestational age-matched non-preeclamptic control and EOPE placentas. Importantly, BMP2 levels were significantly reduced in EOPE placentas compared with controls, and RNAscope in situ hybridization further demonstrated BMP2 expression was disrupted in EOPE placental villi. To explore the molecular mechanisms of BMP2-regulated early trophoblast differentiation, we examined BMP2 expression in first-trimester human placenta and found it to be localized to all subtypes of trophoblasts and the decidua. RNA-seq analysis on control and BMP2-treated primary human trophoblast cells identified 431 differentially expressed genes, including several canonical TGF-ß/BMP signaling targets (BAMBI, ID1, INHBA, IGFBP3). Gene ontology annotations revealed that differentially expressed genes were involved in cell adhesion and extracellular matrix organization. Furthermore, we identified adhesion molecule with IgG-like domain 2 (AMIGO2) as a novel target for BMP2 that contributed to BMP2-induced trophoblast invasion and endothelial-like tube formation. Overall, our findings provide insight into the molecular processes controlled by BMP2 during early placental development that may contribute to the pathogenesis of EOPE.

18.
Sci Rep ; 11(1): 316, 2021 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-33431976

RESUMO

Maternal metabolism dysregulation during pregnancy predisposes offspring to major diseases, including hypertension, in later life, but the mechanism involved remains to be fully elucidated. A high-fat-diet (HFD) pregnant rat model was used to investigate whether excessive intrauterine lipid exposure was associated with elevated blood pressure in offspring and increased levels of leptin, an important biomarker and mediator of vascular dysfunction and hypertension. We found that gestational hyperlipidemia predisposed offspring to blood pressure elevation and sustained increases in leptin levels with no difference in body weight in the rat model. Increased leptin expression and leptin promoter hypomethylation were found in adipose tissues of HFD-exposed offspring. The treatment of mesenchymal stem cells with free fatty acids during adipogenic differentiation resulted in increased leptin expression, accompanied by leptin promoter hypomethylation. In addition, we also followed up 121 children to evaluate the association between maternal triglyceride levels and offspring blood pressure. Consistent with the animal study results, we observed elevated serum leptin levels and blood pressure in the offspring born to women with gestational hypertriglyceridemia. Our findings provide new insights that maternal hyperlipidemia is associated with elevated blood pressure in offspring and is associated with increases in leptin levels through epigenetic memory.


Assuntos
Pressão Sanguínea/genética , Dieta Hiperlipídica/efeitos adversos , Epigênese Genética , Leptina/metabolismo , Exposição Materna/efeitos adversos , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Adiponectina/metabolismo , Animais , Glicemia/metabolismo , Peso Corporal , Feminino , Insulina/metabolismo , Leptina/genética , Masculino , Gravidez , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Ratos , Triglicerídeos/sangue
19.
Differentiation ; 77(2): 162-71, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19281776

RESUMO

We studied the roles of three HOXA genes in cultured normal ovarian surface epithelial (OSE) cells and ovarian cancer cells. They included HOXA4 and HOXA7 because, by cDNA microarray analysis, these were more highly expressed in invasive ovarian carcinomas than in benign or borderline (noninvasive) ovarian tumors, and HOXA9 because it characterizes normal oviductal epithelium, which resembles ovarian serous adenocarcinomas. The three HOXA genes were more highly expressed when OSE cells were dividing and motile than when they were confluent and stationary, and also when they dispersed in response to EGF treatment or to reduced calcium concentrations in culture media. The expression of the HOXA genes varied among ovarian cancer cell lines, but was highest in lines with compact epithelial morphologies. We focused on HOXA4 as the most highly expressed in the ovarian carcinoma array. HOXA4 expression did not parallel proliferative activities of either OSE or ovarian cancer lines. Moreover, modifying HOXA4 expression in ovarian cancer cell lines did not alter either E-cadherin expression or CA125 secretion. However, HOXA4 downregulation enhanced EGFR phosphorylation and migration in serum-starved OSE and ovarian cancer cells in response to EGF, and enhanced migration of all ovarian cancer lines in 5% serum even without EGF treatment. Thus, HOXA4 expression does not correlate with proliferation or with epithelial differentiation, but it increases in response to OSE cell dispersion and negatively regulates EGFR activation and the motility of OSE and of ovarian cancer cells. HOXA4 expression was highest in cancer lines with compact epithelial growth patterns, suggesting, again, an anti-dispersion function. In summary, increased HOXA4 expression in ovarian cancer appears to constitute a tumor-suppressive, homeostatic response to aberrant cell behavior, and, in particular, to cell dispersion and migration.


Assuntos
Células Epiteliais/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Ovarianas/metabolismo , Adulto , Western Blotting , Diferenciação Celular , Linhagem Celular Tumoral , Células Cultivadas , Regulação para Baixo , Feminino , Regulação da Expressão Gênica , Humanos , Pessoa de Meia-Idade , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Epiteliais e Glandulares/fisiopatologia , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/fisiopatologia , Ovário/citologia , Ovário/metabolismo , Padrões de Referência , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Front Cell Dev Biol ; 8: 573781, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33195207

RESUMO

Growth differentiation factor 8 (GDF8) and its antagonist follistatin-like 3 (FSTL3) are expressed in the placenta during early pregnancy. These two factors may have a role to play in the regulation of normal placentation. However, whether GDF8 can regulate the expression of FSTL3 in human trophoblasts remains to be elucidated. In this study, we aimed to investigate the effects of GDF8 on the expression of FSTL3 and the underlying molecular mechanisms using human trophoblasts as a study model. Our results showed that GDF8 significantly upregulates the expression and production of FSTL3, which further promotes cell invasiveness in immortalized extravillous cytotrophoblast cells and primary extravillous cytotrophoblast cells obtained from human first-trimester placentae. Additionally, using an siRNA-mediated knockdown approach, we found that this regulatory effect is most likely mediated by the ALK5-Sma- and Mad-related protein (SMAD)2/3-induced signaling pathway. These findings deepen our understanding of the functional roles of GDF8 and FSTL3 in the regulation of cell invasiveness of trophoblasts.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA