Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Mol Hum Reprod ; 28(10)2022 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-35993908

RESUMO

During placentation, placental cytotrophoblast (CT) cells differentiate into syncytiotrophoblast (ST) cells and extravillous trophoblast (EVT) cells. In the placenta, the expression of various genes is regulated by the Hippo pathway through a transcription complex, Yes-associated protein (YAP)/transcriptional coactivator with PDZ-binding motif (TAZ)-TEA domain transcription factor (TEAD) (YAP/TAZ-TEAD) activity. YAP/TAZ-TEAD activity is controlled by multiple factors and signaling, such as cAMP signaling. cAMP signaling is believed to be involved in the regulation of trophoblast function but is not yet fully understood. Here we showed that YAP/TAZ-TEAD expressions and their activities were altered by cAMP stimulation in BeWo cells, a human choriocarcinoma cell line. The repression of YAP/TAZ-TEAD activity induced the expression of ST-specific genes without cAMP stimulation, and transduction of constitutively active YAP, i.e. YAP-5SA, resulted in the repression of 8Br-cAMP-induced expressions of ST-specific genes in a TEAD-dependent manner. We also investigated the role of YAP/TAZ-TEAD in maintaining CT cells and their differentiation into ST and EVT cells using human trophoblast stem (TS) cells. YAP/TAZ-TEAD activity was involved in maintaining the stemness of TS cells. Induction or repression of YAP/TAZ-TEAD activity resulted in marked changes in the expression of ST-specific genes. Using primary CT cells, which spontaneously differentiate into ST-like cells, the effects of YAP-5SA transduction were investigated, and the expression of ST-specific genes was found to be repressed. These results indicate that the inhibition of YAP/TAZ-TEAD activity, with or without cAMP stimulation, is essential for the differentiation of CT cells into ST cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Trofoblastos , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Feminino , Humanos , Placenta/metabolismo , Gravidez , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Trofoblastos/metabolismo , Proteínas de Sinalização YAP
2.
Endocr J ; 67(1): 91-94, 2020 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-31534059

RESUMO

Uterine sarcomas are rare and aggressive gynecologic tumors with poor prognosis; therefore, early diagnosis is crucial for therapy. However, it is very difficult to distinguish uterine sarcomas from leiomyomas which are common benign uterine tumors. Therefore, the development of a diagnostic method that utilizes reliable biomarkers to distinguish uterine sarcomas from leiomyomas is important so as to identify the rare tumors. The candidate factors as novel biomarkers were searched for in public databases and a pilot study was performed for confirmation. Growth differentiation factor-15 (GDF15), progranulin, and osteopontin were identified as candidate biomarkers for diagnosing uterine sarcoma. Thus, developing a rapid and easy method to measure these factors could help establish a screening system for uterine sarcomas. In this study, we developed a novel measurement system for these factors using a compact chemical luminescence immunological automatic analyzer POCubeTM. This assay system, which is based on the flow-through membrane immunoassay, completes the whole process and generates results within 15 min. Serum concentrations of these factors measured via POCubeTM correlated well with those measured using enzyme-linked immunosorbent assay (r = 0.994 for GDF15, r = 0.992 for progranulin, and r = 0.976 for osteopontin). The POCubeTM system provides rapid and easy measurement of these factors, thereby facilitating uterine sarcoma diagnosis.


Assuntos
Fator 15 de Diferenciação de Crescimento/sangue , Leiomioma/sangue , Osteopontina/sangue , Progranulinas/sangue , Sarcoma/sangue , Neoplasias Uterinas/sangue , Diagnóstico Diferencial , Feminino , Humanos , Imunoensaio , Leiomioma/diagnóstico , Projetos Piloto , Curva ROC , Sarcoma/diagnóstico , Sensibilidade e Especificidade , Fatores de Tempo , Neoplasias Uterinas/diagnóstico
3.
Mol Reprod Dev ; 86(7): 786-797, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31087493

RESUMO

Cyclooxygenase 2 (COX-2) is an inducible rate-limiting enzyme for prostanoid production. Because COX-2 represents one of the inducible genes in mouse mesenchymal stem cells upon differentiation into Leydig cells, we investigated COX-2 expression and production of prostaglandin (PG) in Leydig cells. Although COX-2 was undetectable in mouse testis, it was transiently induced in Leydig cells by human chorionic gonadotropin (hCG) administration. Consistent with the finding that Leydig cells expressed aldo-keto reductase 1B7 (PGF synthase) and PGE synthase 2, induction of COX-2 by hCG caused a marked increase in testicular PGF 2α and PGE 2 levels. Using mouse Leydig cell tumor-derived MA-10 cells as a model, it was indicated by reporter assays and electron mobility shift assays that transcription of the COX-2 gene was activated by CCAAT/enhancer-binding protein ß (C/EBPß) with cAMP-stimulation. C/EBPß expression was induced by cAMP-stimulation, whereas expression of C/EBP homolog protein (CHOP) was robustly downregulated. Transfection of CHOP expression plasmid inhibited cAMP-induced COX-2 promoter activity. In addition, CHOP reduced constitutive COX-2 expression in other mouse Leydig cell tumor-derived TM3 cells. These results indicate that COX-2 is induced in Leydig cells by activation of C/EBPß via reduction of CHOP expression upon gonadotropin-stimulation to produce PGF 2α and PGE 2 .


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Gonadotropina Coriônica/farmacologia , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Células Intersticiais do Testículo/metabolismo , Substâncias para o Controle da Reprodução/farmacologia , Animais , Linhagem Celular Tumoral , AMP Cíclico/metabolismo , Ciclo-Oxigenase 2/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição CHOP/genética , Fator de Transcrição CHOP/metabolismo , Transcrição Gênica , Transfecção
4.
Endocr J ; 63(11): 943-951, 2016 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-27681884

RESUMO

Steroid hormones are mainly produced in adrenal glands and gonads. Because steroid hormones play vital roles in various physiological processes, replacement of deficient steroid hormones by hormone replacement therapy (HRT) is necessary for patients with adrenal and gonadal failure. In addition to HRT, tissue regeneration using stem cells is predicted to provide novel therapy. Among various stem cell types, mesenchymal stem cells can be differentiated into steroidogenic cells following ectopic expression of nuclear receptor (NR) 5A subfamily proteins, steroidogenic factor-1 (also known as adrenal 4 binding protein) and liver receptor homolog-1, with the aid of cAMP signaling. Conversely, these approaches cannot be applied to pluripotent stem cells, such as embryonic stem cells and induced pluripotent stem cells, because of poor survival following cytotoxic expression of NR5A subfamily proteins. However, if pluripotent stem cells are first differentiated through mesenchymal lineage, they can also be differentiated into steroidogenic cells via NR5A subfamily protein expression. This approach offers a potential suitable cells for future regenerative medicine and gene therapy for diseases caused by steroidogenesis deficiencies. It represents a powerful tool to investigate the molecular mechanisms involved in steroidogenesis. This article highlights our own and current research on the induction of steroidogenic cells from various stem cells. We also discuss the future direction of their clinical application.


Assuntos
Células-Tronco Adultas/fisiologia , Hormônios/biossíntese , Células-Tronco Pluripotentes/fisiologia , Esteroides/biossíntese , Engenharia Tecidual/métodos , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Animais , Técnicas de Cultura de Células/métodos , Diferenciação Celular/fisiologia , Terapia Genética , Terapia de Reposição Hormonal , Humanos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Engenharia Tecidual/tendências
5.
Biochim Biophys Acta ; 1839(1): 33-42, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24321386

RESUMO

Ferredoxin reductase (FDXR, also known as adrenodoxin reductase) is a mitochondrial flavoprotein that transfers electrons from NADPH to mitochondrial cytochrome P450 enzymes, mediating the function of an iron-sulfur cluster protein, ferredoxin. FDXR functions in various metabolic processes including steroidogenesis. It is well known that multiple steroidogenic enzymes are regulated by a transcription factor steroidogenic factor-1 (SF-1, also known as Ad4BP). Previously, we have shown that SF-1 transduction causes human mesenchymal stem cell differentiation into steroidogenic cells. Genome-wide analysis of differentiated cells, using a combination of DNA microarray and promoter tiling array analyses, showed that FDXR is a novel SF-1 target gene. In this study, the transcriptional regulatory mechanism of FDXR was examined in steroidogenic cells. A chromatin immunoprecipitation assay revealed that a novel SF-1 binding region was located within intron 2 of the human FDXR gene. Luciferase reporter assays showed that FDXR transcription was activated through the novel SF-1 binding site within intron 2. Endogenous SF-1 knockdown in human adrenocortical H295R and KGN cells decreased FDXR expression. In H295R cells, strong binding of two histone markers of active enhancers, histones H3K27ac and H3K4me2, were detected near the SF-1 binding site within intron 2. Furthermore, the binding of these histone markers was decreased concurrent with SF-1 knockdown in H295R cells. These results indicated that abundant FDXR expression in these steroidogenic cells was maintained through SF-1 binding to the intronic enhancer of the FDXR gene.


Assuntos
Elementos Facilitadores Genéticos , Ferredoxina-NADP Redutase/genética , Fator Esteroidogênico 1/genética , Esteroides/metabolismo , Transcrição Gênica , Sítios de Ligação , Linhagem Celular , Proteínas de Ligação a DNA , Ferredoxina-NADP Redutase/metabolismo , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Histonas/genética , Humanos , Íntrons , Histona Desmetilases com o Domínio Jumonji/genética , Sequências Reguladoras de Ácido Nucleico , Fator Esteroidogênico 1/metabolismo , Esteroides/biossíntese
6.
Biochim Biophys Acta ; 1839(5): 406-14, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24705138

RESUMO

Pluripotent stem cells maintain their pluripotency and undifferentiated status through a network of transcription factors. Liver receptor homolog-1 (Lrh-1) is one of these, and regulates the expression of other important transcription factors such as Oct-3/4 and Nanog. In early embryo and embryonic stem (ES) cells, Lrh-1 is transcribed using a unique promoter. In this study, we investigated the transcriptional regulation of embryonic Lrh-1 using ES and embryonal carcinoma F9 cells. Reporter assays, electrophoretic mobility shift assays, and chromatin immunoprecipitation assays demonstrated that Sox2 and Gabp proteins bind to the promoter region of embryonic Lrh-1, and are necessary for its activation. The Sox2 site showed strong promoter activity and affinity for protein binding. Upon differentiation into the parietal endoderm by retinoic acid and cAMP, Lrh-1 promoter activity and transcripts were markedly reduced within 24 h. At the same time, Sox2 and Gabp binding to the promoter region of Lrh-1 were decreased, followed by a reduction of their expression. These results indicate that embryonic Lrh-1 expression is regulated by both Sox2 and Gabp. Our study presents new insights into the transcription factor network of pluripotent stem cells.


Assuntos
Células-Tronco Embrionárias/fisiologia , Fator de Transcrição de Proteínas de Ligação GA/genética , Receptores Citoplasmáticos e Nucleares/genética , Fatores de Transcrição SOXB1/genética , Animais , Sequência de Bases , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Fator de Transcrição de Proteínas de Ligação GA/metabolismo , Regulação da Expressão Gênica , Camundongos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/fisiologia , Regiões Promotoras Genéticas , Ligação Proteica , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Transcrição Gênica , Transfecção
7.
Zoolog Sci ; 32(4): 323-30, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26245218

RESUMO

Steroidogenic factor-1 (SF-1) and liver receptor homolog-1 (LRH-1) belong to the nuclear receptor superfamily and are categorized as orphan receptors. In addition to other nuclear receptors, these play roles in various physiological phenomena by regulating the transcription of target genes. Both factors share very similar structures and exhibit common functions. Of these, the roles of SF-1 and LRH-1 in steroidogenesis are the most important, especially that of SF-1, which was originally discovered and named to reflect such roles. SF-1 and LRH-1 are essential for steroid hormone production in gonads and adrenal glands through the regulation of various steroidogenesis-related genes. As SF-1 is also necessary for the development of gonads and adrenal glands, it is also considered a master regulator of steroidogenesis. Recent studies have clearly demonstrated that LRH-1 also represents another master regulator of steroidogenesis, which similarly to SF-1, can induce differentiation of non-steroidogenic stem cells into steroidogenic cells. Here, we review the functions of both factors in these steroidogenesis-related phenomena.


Assuntos
Receptores Citoplasmáticos e Nucleares/fisiologia , Fator Esteroidogênico 1/metabolismo , Esteroides/biossíntese , Animais , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Fator Esteroidogênico 1/genética , Esteroides/metabolismo
8.
Endocr J ; 62(9): 757-63, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26135521

RESUMO

Steroid hormones are synthesized from cholesterol in various tissues, mainly in the adrenal glands and gonads. Because these lipid-soluble steroid hormones immediately diffuse through the cells in which they are produced, their secretion directly reflects the activity of the genes related to their production. Progesterone is important not only for luteinization and maintenance of pregnancy, but also as a substrate for most other steroids. Steroidogenic acute regulatory protein (STAR), cytochrome P450 cholesterol side-chain cleavage enzyme (P450scc), and 3ß-hydroxysteroid dehydrogenase/Δ(5)-Δ(4) isomerase (3ß-HSD) are well-known proteins essential for progesterone production. In addition to them, glutathione S-transferase A1-1 and A3-3 are shown to exert Δ(5)-Δ(4) isomerization activity to produce progesterone in a cooperative fashion with 3ß-HSD. 5-Aminolevulinic acid synthase 1, ferredoxin 1, and ferredoxin reductase also play a role in steroidogenesis as accessory factors. Members of the nuclear receptor 5A (NR5A) family (steroidogenic factor 1 and liver receptor homolog 1) play a crucial role in the transcriptional regulation of these genes. The NR5A family activates these genes by binding to NR5A responsive elements present within their promoter regions, as well as to the elements far from their promoters. In addition, various NR5A-interacting proteins including peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), nuclear receptor subfamily 0, group B, member 1 (DAX-1), and CCAAT/enhancer-binding proteins (C/EBP) are involved in the transcription of NR5A target genes and regulate the transcription either positively or negatively under both basal and tropic hormone-stimulated conditions. In this review, we describe the transcriptional regulation of genes related to progesterone production.


Assuntos
Regulação da Expressão Gênica , Progesterona/biossíntese , Transcrição Gênica , 17-Hidroxiesteroide Desidrogenases/genética , Enzima de Clivagem da Cadeia Lateral do Colesterol/genética , Glutationa Transferase/genética , Humanos , Fosfoproteínas/genética , Regiões Promotoras Genéticas , Fator Esteroidogênico 1/genética
9.
Biochem J ; 460(3): 459-71, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24635384

RESUMO

The transcription factor SF-1 (steroidogenic factor-1) is a master regulator of steroidogenesis. Previously, we have found that SF-1 induces the differentiation of mesenchymal stem cells into steroidogenic cells. To elucidate the molecular mechanisms of SF-1-mediated functions, we attempted to identify protein components of the SF-1 nuclear protein complex in differentiated cells. SF-1 immunoaffinity chromatography followed by MS/MS analysis was performed, and 24 proteins were identified. Among these proteins, we focused on C/EBPß (CCAAT/enhancer-binding protein ß), which is an essential transcription factor for ovulation and luteinization, as the transcriptional mechanisms of C/EBPß working together with SF-1 are poorly understood. C/EBPß knockdown attenuated cAMP-induced progesterone production in granulosa tumour-derived KGN cells by altering STAR (steroidogenic acute regulatory protein), CYP11A1 (cytochrome P450, family 11, subfamily A, polypeptide 1) and HSD3B2 (hydroxy-δ-5-steroid dehydrogenase, 3ß- and steroid δ-isomerase 2) expression. EMSA and ChIP assays revealed novel C/EBPß-binding sites in the upstream regions of the HSD3B2 and CYP11A1 genes. These interactions were enhanced by cAMP stimulation. Luciferase assays showed that C/EBPß-responsive regions were found in each promoter and C/EBPß is involved in the cAMP-induced transcriptional activity of these genes together with SF-1. These results indicate that C/EBPß is an important mediator of progesterone production by working together with SF-1, especially under tropic hormone-stimulated conditions.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Progesterona/biossíntese , Fator Esteroidogênico 1/fisiologia , Animais , Proteína beta Intensificadora de Ligação a CCAAT/genética , Células Cultivadas , Enzima de Clivagem da Cadeia Lateral do Colesterol/genética , Regulação da Expressão Gênica , Humanos , Camundongos , Fosfoproteínas , Progesterona/genética , Progesterona Redutase/genética , Espectrometria de Massas em Tandem
10.
FASEB J ; 27(8): 3198-208, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23650189

RESUMO

Steroidogenic factor 1 (SF-1) is a master regulator for steroidogenesis. In this study, we identified novel SF-1 target genes using a genome-wide promoter tiling array and a DNA microarray. SF-1 was found to regulate human glutathione S-transferase A (GSTA) family genes (hGSTA1-hGSTA4), a superfamily of detoxification enzymes clustered on chromosome 6p12. All hGSTA genes were up-regulated by transduction of SF-1 into human mesenchymal stem cells, while knockdown of endogenous SF-1 in H295R cells down-regulated all hGSTA genes. Chromatin immunoprecipitation assays, however, revealed that SF-1 bound directly to the promoters of hGSTA3 and weakly of hGSTA4. Chromosome conformation capture assays revealed that the coordinated expression of the genes was based on changes in higher-order chromatin structure triggered by SF-1, which enables the formation of long-range interactions, at least between hGSTA1 and hGSTA3 gene promoters. In steroidogenesis, dehydrogenation of the 3-hydroxy group and subsequent Δ(5)-Δ(4) isomerization are thought to be enzymatic properties of 3ß-hydroxysteroid dehydrogenase (3ß-HSD). Here, we demonstrated that, in steroidogenic cells, the hGSTA1 and hGSTA3 gene products catalyze Δ(5)-Δ(4) isomerization in a coordinated fashion with 3ß-HSD II to produce progesterone or Δ(4)-androstenedione from their Δ(5)-precursors. Thus, hGSTA1 and hGSTA3 gene products are new members of steroidogenesis working as Δ(5)-Δ(4) isomerases.


Assuntos
Glutationa Transferase/metabolismo , Isoenzimas/metabolismo , Fator Esteroidogênico 1/metabolismo , Esteroides/biossíntese , Androstenodiona/biossíntese , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Glutationa Transferase/síntese química , Glutationa Transferase/genética , Humanos , Isoenzimas/genética , Células-Tronco Mesenquimais/metabolismo , Mutação , Análise de Sequência com Séries de Oligonucleotídeos , Progesterona/biossíntese , Progesterona Redutase/genética , Progesterona Redutase/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator Esteroidogênico 1/genética
11.
J Biol Chem ; 285(36): 28240-51, 2010 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-20601698

RESUMO

StAR (steroidogenic acute regulatory protein) mediates the transport of cholesterol from the outer to the inner mitochondrial membrane, the process of which is the rate-limiting step for steroidogenesis. Transcriptional regulation of the proximal promoter of the human StAR gene has been well characterized, whereas analysis of its distal control region has not. Recently, we found that SF-1 (steroidogenic factor 1) induced the differentiation of mesenchymal stem cells (MSCs) into steroidogenic cells with the concomitant strong induction of StAR expression. Here, we show, using differentiated MSCs, that StAR expression is regulated by a novel distal control region. Using electrophoretic mobility shift (EMSA) and chromatin immunoprecipitation (ChIP) assays, we identified novel SF-1 binding sites between 3,000 and 3,400 bp upstream of StAR. A luciferase reporter assay revealed that the region worked as a strong regulator to exert maximal transcription of StAR. ChIP analysis of histone H3 revealed that upon SF-1 expression, nucleosome eviction took place at the SF-1 binding sites, not only in the promoter but also in the distal SF-1 binding sites. Chromosome conformation capture analysis revealed that the region upstream of StAR formed a chromatin loop both in the differentiated MSCs and in KGN cells, a human granulosa cell tumor cell line, where SF-1 is endogenously expressed. Finally, SF-1 knockdown resulted in disrupted formation of this chromatin loop in KGN cells. These results indicate that the novel distal control region participate in StAR activation through SF-1 dependent alterations of chromatin structure, including histone eviction and chromatin loop formation.


Assuntos
Cromatina/metabolismo , Região de Controle de Locus Gênico/genética , Fosfoproteínas/genética , Fator Esteroidogênico 1/metabolismo , Animais , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Sequência Conservada , Ensaio de Desvio de Mobilidade Eletroforética , Evolução Molecular , Histonas/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Dados de Sequência Molecular , Fosfoproteínas/metabolismo , Ratos , Ativação Transcricional
12.
Gen Comp Endocrinol ; 170(1): 207-12, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21036175

RESUMO

In certain species of amphibians gonadal differentiation is influenced by steroid hormones. In the case of the frog Rana rugosa testosterone given to tadpoles reverses sex from female to male, while the opposite reversal - male to female - can be achieved using estradiol-17ß. In this study, we investigated whether CYP19 (P450 aromatase), the enzyme responsible for a production of estradiol-17ß, was present in the differentiating gonad of R. rugosa. Initially, we immunized rabbits against frog CYP19 peptides and performed immunostaining using specific antibodies purified from that serum. CYP19-reactive signals were observed in gonadal somatic cells of the female, but not male tadpoles at stage (St.) I (the stage prior to phenotypic sex determination in tadpoles of R. rugosa). Immunopositive signals were also produced in ovarian somatic cells froglets at St. XXV (just after the completion of metamorphosis). We also examined the enzymatic activity of CYP19 in the differentiating gonad of R. rugosa. Reverse-phase HPLC (high performance liquid chromatography) analysis revealed that [(3)H]testosterone was converted to [(3)H]estradiol-17ß in the gonad of tadpoles at St. I. Interestingly, the rate of conversion was much higher in females than in males. To the best of our knowledge, this is the first report on the biosynthesis of estradiol-17ß in the gonad of amphibians, and the co-incident identification of active CYP19 enzyme in the differentiating gonad of R. rugosa. Based on our results, we conclude that estradiol-17ß may be involved in ovarian differentiation in this species.


Assuntos
Estradiol/biossíntese , Gônadas/metabolismo , Ranidae/metabolismo , Animais , Aromatase/metabolismo , Western Blotting , Cromatografia Líquida de Alta Pressão , Estradiol/metabolismo , Feminino , Imuno-Histoquímica , Masculino
13.
Biomed Res Int ; 2019: 8973076, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31058195

RESUMO

Ovaries represent one of the primary steroidogenic organs, producing estrogen and progesterone under the regulation of gonadotropins during the estrous cycle. Gonadotropins fluctuate the expression of various steroidogenesis-related genes, such as those encoding steroidogenic enzymes, cholesterol deliverer, and electronic transporter. Steroidogenic factor-1 (SF-1)/adrenal 4-binding protein (Ad4BP)/NR5A1 and liver receptor homolog-1 (LRH-1) play important roles in these phenomena via transcriptional regulation. With the aid of cAMP, SF-1/Ad4BP and LRH-1 can induce the differentiation of stem cells into steroidogenic cells. This model is a useful tool for studying the molecular mechanisms of steroidogenesis. In this article, we will provide insight into the transcriptional regulation of steroidogenesis-related genes in ovaries that are revealed from stem cell-derived steroidogenic cells. Using the cells derived from the model, novel SF-1/Ad4BP- and LRH-1-regulated genes were identified by combined DNA microarray and promoter tiling array analyses. The interaction of SF-1/Ad4BP and LRH-1 with transcriptional regulators in the regulation of ovarian steroidogenesis was also revealed.


Assuntos
Ovário/crescimento & desenvolvimento , Receptores Citoplasmáticos e Nucleares/genética , Fator Esteroidogênico 1/genética , Transcrição Gênica , Diferenciação Celular/genética , Feminino , Regulação da Expressão Gênica/genética , Humanos , Ovário/metabolismo , Regiões Promotoras Genéticas , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Transcrição/genética
14.
Endocrinology ; 149(4): 1786-92, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18162527

RESUMO

We have shown previously that Cyp11b1, an 11beta-hydroxylase responsible for glucocorticoid biosynthesis in the adrenal gland, was induced by cAMP in androgen-producing Leydig-like cells derived from mesenchymal stem cells. We found that Cyp11b1 was induced in male Leydig cells, or female theca cells, when human chorionic gonadotropin was administered in immature mice. Expression of Cyp11b1 in rodent gonads caused the production of 11-ketotestosterone (11-KT), a major fish androgen, which induces male differentiation or spermatogenesis in fish. As in teleosts, plasma concentrations of 11-KT were elevated in human chorionic gonadotropin-treated mice. In contrast to teleosts, however, plasma concentrations of 11-KT were similar in both sexes, despite levels of testosterone, a precursor substrate, being about 20 times higher in male mice. Because expression of 11beta-hydroxysteroid dehydrogenase type 2, was much higher in the mouse ovary than in the testis, conversion of testosterone into 11-KT may occur more efficiently in the ovary. In a luciferase reporter system that was responsive to and activated by androgens, 11-KT efficiently activated mammalian androgen receptor-mediated transactivation. Our results suggest that the androgen metabolic pathway is conserved between teleosts and mammals, despite sexual dominance and reproductive functions of 11-KT being altered during evolution.


Assuntos
Androgênios/metabolismo , Gonadotropina Coriônica/farmacologia , Gônadas/metabolismo , Hormônio Luteinizante/farmacologia , Esteroide 11-beta-Hidroxilase/biossíntese , Testosterona/análogos & derivados , 11-beta-Hidroxiesteroide Desidrogenases/fisiologia , Animais , Evolução Biológica , Células Cultivadas , Indução Enzimática , Camundongos , Camundongos Endogâmicos C57BL , Receptores Androgênicos/metabolismo , Testosterona/biossíntese
15.
Endocrinology ; 149(4): 1524-33, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18174289

RESUMO

Estrogen has been considered to enhance FSH actions in the ovary, including the induction of the LH receptor (LHR). In this study, we elucidated the mechanism underlying the effect of estrogen on the induction of LHR by FSH in rat granulosa cells. Estradiol clearly enhanced the FSH-induced LHR mRNA increase in a time- and dose-dependent manner, with a maximum increase of approximately 3.5-fold at 72 h, compared with the level of LHR mRNA solely induced by FSH. We then investigated whether the effect of estrogen on LHR mRNA was due to increased transcription and/or altered mRNA stability. A luciferase assay with the plasmid containing the LHR 5'-flanking region did not show that estradiol increased the promoter activity induced by FSH. In contrast, the decay curves for LHR mRNA showed a significant increase in half-life with FSH and estradiol, suggesting that the increased stability of LHR mRNA is at least responsible for the regulation of LHR mRNA by estrogen. Recently mevalonate kinase (Mvk) was identified as a trans-factor that binds to LHR mRNA and alters LHR mRNA stability in the ovary. We found that estradiol, with FSH, decreased Mvk mRNA levels in rat granulosa cell culture, resulting in up-regulation of LHR mRNA that was inversely correlated to Mvk mRNA expression. Furthermore, the augmentation of FSH-induced LHR expression in the presence of estrogen was erased with the overexpression of Mvk by transient transfection. Taken together, these data indicate that LHR mRNA is up-regulated due to increased stability when estrogen negatively controls Mvk.


Assuntos
Estradiol/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Células da Granulosa/metabolismo , RNA Mensageiro/análise , Receptores do LH/genética , Animais , Células Cultivadas , Feminino , Hormônio Foliculoestimulante/farmacologia , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Ratos , Ratos Wistar
16.
Mol Reprod Dev ; 75(5): 931-9, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18163434

RESUMO

NR4A1, also called NGFI-B in the rat, Nur77 in the mouse and TR3 in humans, belongs to the orphan nuclear steroid hormone receptor superfamily and is one of the immediate-early genes. In the endocrine organs, including the gonads, NGFI-B/Nur77 gene expression is rapidly induced by pituitary hormones. NGFI-B/Nur77 expression was found to be rapidly reduced by an estrogenic endocrine disrupter, diethylstilbestrol (DES) in theca interna cells of immature rat ovaries. DES treatment also triggered a rapid decrease of serum luteinizing hormone (LH) levels, suggesting that DES acts on the hypothalamo-pituitary axis to suppress LH secretion from the pituitary. The transcriptional regulation of NGFI-B/Nur77 by LH/human chorionic gonadotropin (hCG) or 8-bromoadenosine 3'-5'-cyclic monophosphate (8 Br-cAMP) was examined in mouse Leydig tumor cells MA-10. Luciferase assays using NGFI-B/Nur77 promoter constructs and electric mobility shift assays (EMSA) showed that NGFI-B/Nur77 gene expression was mediated through three of the four activator protein-1 (AP-1)-like sites, namely the -233 AP-1, -213 AP-1 and -69 AP-1 sites adjacent to the transcription start site of the NGFI-B/Nur77 promoter. We also demonstrated here that both the Jun family and cAMP-responsive element binding (CREB) proteins bind to the -233 AP-1 site, whereas the main binding protein to the -213 AP-1 site was CREB, and Jun family protein to the -69 AP-1 site, respectively. The rapid induction of NGFI-B/Nur77 gene expression by LH/hCG in MA-10 cells appears to be mediated by both CREB and Jun family proteins through the cAMP-protein kinase A (PKA) pathway.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Regulação Neoplásica da Expressão Gênica , Tumor de Células de Leydig/metabolismo , Receptores de Esteroides/biossíntese , Células Tecais/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Animais , Linhagem Celular Tumoral , Gonadotropina Coriônica/farmacologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Dietilestilbestrol/farmacologia , Estrogênios não Esteroides/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Sistema Hipotálamo-Hipofisário/metabolismo , Tumor de Células de Leydig/patologia , Hormônio Luteinizante/farmacologia , Camundongos , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares , Proteínas Proto-Oncogênicas c-jun/metabolismo , Ratos , Elementos de Resposta , Células Tecais/patologia , Fator de Transcrição AP-1
17.
Reprod Biol Endocrinol ; 6: 62, 2008 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-19077323

RESUMO

BACKGROUND: P450 oxidoreductase (POR) catalyzes electron transfer to microsomal P450 enzymes. Its deficiency causes Antley-Bixler syndrome (ABS), and about half the patients with ABS have ambiguous genitalia and/or impaired steroidogenesis. POR mRNA expression is up-regulated when mesenchymal stem cells (MSCs) differentiate into steroidogenic cells, suggesting that the regulation of POR gene expression is important for steroidogenesis. In this context we examined the regulation of POR expression in ovarian granulosa cells by gonadotropins, and its possible role in steroidogenesis. METHODS: Changes in gene expression in MSCs during differentiation into steroidogenic cells were examined by DNA microarray analysis. Changes in mRNA and protein expression of POR in the rat ovary or in granulosa cells induced by gonadotropin treatment were examined by reverse transcription-polymerase chain reaction and western blotting. Effects of transient expression of wild-type or mutant (R457H or V492E) POR proteins on the production of estrone in COS-7 cells were examined in vitro. Effects of POR knockdown were also examined in estrogen producing cell-line, KGN cells. RESULTS: POR mRNA was induced in MSCs following transduction with the SF-1 retrovirus, and was further increased by cAMP treatment. Expression of POR mRNA, as well as Cyp19 mRNA, in the rat ovary were induced by equine chorionic gonadotropin and human chorionic gonadotropin. POR mRNA and protein were also induced by follicle stimulating hormone in primary cultured rat granulosa cells, and the induction pattern was similar to that for aromatase. Transient expression of POR in COS-7 cells, which expressed a constant amount of aromatase protein, greatly increased the rate of conversion of androstenedione to estrone, in a dose-dependent manner. The expression of mutant POR proteins (R457H or V492E), such as those found in ABS patients, had much less effect on aromatase activity than expression of wild-type POR proteins. Knockdown of endogenous POR protein in KGN human granulosa cells led to reduced estrone production, indicating that endogenous POR affected aromatase activity. CONCLUSION: We demonstrated that the expression of POR, together with that of aromatase, was regulated by gonadotropins, and that its induction could up-regulate aromatase activity in the ovary, resulting in a coordinated increase in estrogen production.


Assuntos
Estrogênios/biossíntese , Gonadotropinas/fisiologia , Células da Granulosa/enzimologia , NADPH-Ferri-Hemoproteína Redutase/metabolismo , Animais , Aromatase/metabolismo , Células COS , Diferenciação Celular , Linhagem Celular , Chlorocebus aethiops , Estrona/biossíntese , Feminino , Regulação da Expressão Gênica , Células da Granulosa/metabolismo , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/enzimologia , Células-Tronco Mesenquimais/metabolismo , Mutação , NADPH-Ferri-Hemoproteína Redutase/genética , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/metabolismo , Ratos , Fatores de Tempo
18.
Biochem Pharmacol ; 154: 136-147, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29674000

RESUMO

Hypertension is considered as one of the cancer progressive factors, and often found comorbidity in cancer patients. Renin-angiotensin system (RAS) plays an important role in the regulation of blood pressure, and angiotensin II (Ang II) is well known pressor peptide associated with RAS. Ang II has been reported to accelerate progression and metastasis of cancer cells. However, its precise mechanisms have not been fully understood. In this study, we sought to elucidate the mechanisms by which Ang II exacerbates hematogenous metastasis in mouse melanoma cells, focusing the adhesion pathway in vascular endothelial cells. For this purpose, B16/F10 mouse melanoma cells, which do not express the Ang II type 1 receptor (AT1R), were intravenously injected into C57BL/6 mice. Two weeks after cell injection, the number of lung metastatic colonies was significantly higher in the Ang II-treated group (1 µg/kg/min) than in the vehicle-treated group. The AT1R blocker valsartan (40 mg/kg/day), but not the calcium channel blocker amlodipine (5 or 10 mg/kg/day), significantly suppressed the effect of Ang II. In endothelium-specific Agtr1a knockout mice, Ang II-mediated acceleration of lung metastases of melanoma cells was significantly diminished. Ang II treatment significantly increased E-selectin mRNA expression in vascular endothelial cells collected from lung tissues, and thus promoted adherence of melanoma cells to the vascular endothelium. Ang II-accelerated lung metastases of melanoma cells were also suppressed by treatment with anti-E-selectin antibody (20 mg/kg). Taken together, Ang II-treatment exacerbates hematogenous cancer metastasis by promoting E-selectin-mediated adhesion of cancer cells to vascular endothelial cells.


Assuntos
Angiotensina II/toxicidade , Moléculas de Adesão Celular/metabolismo , Células Endoteliais/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Melanoma Experimental/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Relação Dose-Resposta a Droga , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Neoplasias Pulmonares/patologia , Masculino , Melanoma Experimental/induzido quimicamente , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Distribuição Aleatória
19.
Front Biosci ; 12: 1911-26, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17127430

RESUMO

Corepressor BS69 interacts with ZHX1, a member of the ZHX family having zinc-fingers and homeoboxes. In the rat, we have identified four forms of splicing variants, BS69alpha, BS69beta, BS69gamma, and BS69delta. Based on the amino acid sequence, BS69alpha corresponded to the human orthologue. BS69beta and BS69gamma contain a novel 56 amino acid region encoded by the exon 11b of the rat BS69 gene. Both BS69gamma and BS69delta lacked a region encoded by exon 3 of the gene. Although all four variants were ubiquitously expressed in rats, the transcripts having the exon 11b were detected in mice and rats but not in humans. A common C-terminal MYND domain of BS69 was required for the interaction with PxLxP motif of ZHX1. Although BS69 was originally found as a corepressor interacting with ZHX1, BS69 was also found to function as a transcriptional activator in HEK293 cells, in which the activation required the MYND domain of BS69. Co-transfection of BS69 with a mutant form of ZHX1, which cannot interact with BS69, led to increase the transcriptional activation of BS69, suggesting that transcriptional activation mediated by BS69 is suppressed by ZHX1. In contrast, BS69 showed transcriptional repression in COS-7 and CV-1 cells and the repression domain was mapped to the N-terminus of BS69beta. Both the wild type and mutant form of ZHX1 had no effect on the BS69 repression, suggesting that the repression mediated by BS69 in COS-7 and CV-1 cells may require a cofactor other than ZHX1 in the cells. Therefore, our results suggest that BS69 may function either as a transcriptional repressor or as a transcriptional activator depending on its regulatory partner.


Assuntos
Proteínas de Homeodomínio/metabolismo , Proteínas Repressoras/metabolismo , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Processamento Alternativo , Animais , Sítios de Ligação , Células COS , Linhagem Celular , Chlorocebus aethiops , Clonagem Molecular , Proteínas de Homeodomínio/química , Humanos , Camundongos , Estrutura Terciária de Proteína , Ratos , Proteínas Repressoras/química , Proteínas Repressoras/genética , Transativadores/química , Transativadores/genética , Fatores de Transcrição/química , Técnicas do Sistema de Duplo-Híbrido
20.
Sci Rep ; 7(1): 8374, 2017 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-28827713

RESUMO

Diethylstilbestrol (DES), a strong estrogenic compound, is well-known to affect the reproductive system. In this study, we investigated the effects of DES administration on gonadotropin levels and ovarian steroidogenesis in prepubertal rats. DES treatment acutely reduced serum LH levels, followed by a reduction in the expression of various steroidogenesis-related genes in theca cells. Serum FSH levels were almost unaffected by DES-treatment, even though Cyp19a1 expression was markedly reduced. Serum progesterone, testosterone and estradiol levels were also declined at this time. LH levels recovered from 12 h after DES-treatment and gradually increased until 96 h with a reduction of ERα expression observed in the pituitary. Steroidogenesis-related genes were also up-regulated during this time, except for Cyp17a1 and Cyp19a1. Consistent with observed gene expression pattern, serum testosterone and estradiol concentrations were maintained at lower levels, even though progesterone levels recovered. DES-treatment induced the inducible nitric oxide synthase (iNOS) in granulosa cells, and a nitric oxide generator markedly repressed Cyp19a1 expression in cultured granulosa cells. These results indicate that DES inhibits thecal androgen production via suppression of pituitary LH secretion and ovarian Cyp17a1 expression. In addition, DES represses Cyp19a1 expression by inducing iNOS gene expression for continuous inhibition of estrogen production in granulosa cells.


Assuntos
Androgênios/sangue , Aromatase/genética , Dietilestilbestrol/administração & dosagem , Estrogênios não Esteroides/administração & dosagem , Estrogênios/sangue , Células da Granulosa/efeitos dos fármacos , Ovário/efeitos dos fármacos , Células Tecais/efeitos dos fármacos , Animais , Feminino , Perfilação da Expressão Gênica , Gonadotropinas/sangue , Células da Granulosa/metabolismo , Ovário/metabolismo , Ratos , Esteroide 17-alfa-Hidroxilase/análise , Esteroide 17-alfa-Hidroxilase/genética , Células Tecais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA