Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 9(1): 15436, 2019 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-31659170

RESUMO

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

2.
Environ Health Perspect ; 126(4): 047006, 2018 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-29665328

RESUMO

BACKGROUND: Analgesic exposure during pregnancy may affect aspects of fetal gonadal development that are targeted by endocrine disruptors. OBJECTIVES: We investigated whether therapeutically relevant doses of acetaminophen and ibuprofen affect germ cell (GC) development in human fetal testes/ovaries using in vitro and xenograft approaches. METHODS: First-trimester human fetal testes/ovaries were cultured and exposed to acetaminophen or ibuprofen (7 d). Second-trimester human fetal testes were xenografted into mice and exposed to acetaminophen (1 or 7 d), or ibuprofen (7 d). To determine mechanism of action, a human GC tumor­derived cell line (NTera2) exhibiting fetal GC characteristics was used in addition to in vitro and in vivo rat models. RESULTS AND DISCUSSION: Gonocyte (TFAP2C+) number was reduced relative to controls in first-trimester human fetal testes exposed in vitro to acetaminophen (-28%) or ibuprofen (-22%) and also in ovaries exposed to acetaminophen (-43%) or ibuprofen (-49%). Acetaminophen exposure reduced gonocyte number by 17% and 30% in xenografted second-trimester human fetal testes after treatment of host mice for 1 or 7 d, respectively. NTera2 cell number was reduced following exposure to either analgesic or prostaglandin E2 (PGE2) receptor antagonists, whereas PGE2 agonists prevented acetaminophen-induced reduction in NTera2 cell number. Expression of GC pluripotency genes, and genes that regulate DNA/histone methylation, also differed from controls following analgesic and PGE2 receptor antagonist exposures. Gene expression changes were observed in rat fetal testis/ovary cultures and after in vivo acetaminophen exposure of pregnant rats. For example, expression of the epigenetic regulator TET1, was increased following exposure to acetaminophen in human NTera2 cells, rat fetal testis/ovary cultures, and in fetal testes and ovaries after in vivo exposure of pregnant rats, indicating translatability across experimental models and species. CONCLUSIONS: Our results demonstrate evidence of PGE2-mediated effects of acetaminophen and ibuprofen on GC/NTera2 cells, which raises concerns about analgesic use during human pregnancy that warrant further investigation. https://doi.org/10.1289/EHP2307.


Assuntos
Acetaminofen/efeitos adversos , Diferenciação Celular/efeitos dos fármacos , Desenvolvimento Fetal/efeitos dos fármacos , Células Germinativas/efeitos dos fármacos , Ibuprofeno/efeitos adversos , Animais , Relação Dose-Resposta a Droga , Feminino , Xenoenxertos , Humanos , Técnicas In Vitro , Masculino , Camundongos , Camundongos Nus , Ovário/efeitos dos fármacos , Gravidez , Primeiro Trimestre da Gravidez , Segundo Trimestre da Gravidez , Testículo/efeitos dos fármacos
3.
Sci Rep ; 7(1): 2521, 2017 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-28566680

RESUMO

Administration of dibutyl phthalate (DBP) to pregnant rats causes reproductive disorders in male offspring, resulting from suppression of intratesticular testosterone, and is used as a model for human testicular dysgenesis syndrome (TDS). DBP exposure in pregnancy induces focal dysgenetic areas in fetal testes that appear between e19.5-e21.5, manifesting as focal aggregation of Leydig cells and ectopic Sertoli cells (SC). Our aim was to identify the origins of the ectopic SC. Time-mated female rats were administered 750 mg/kg/day DBP in three different time windows: full window (FW; e13.5-e20.5), masculinisation programming window (MPW; e15.5-e18.5), late window (LW; e19.5-e20.5). We show that DBP-MPW treatment produces more extensive and severe dysgenetic areas, with more ectopic SC and germ cells (GC) than DBP-FW treatment; DBP-LW induces no dysgenesis. Our findings demonstrate that ectopic SC do not differentiate de novo, but result from rupture of normally formed seminiferous cords beyond e20.5. The more severe testis dysgenesis in DBP-MPW animals may result from the presence of basally migrating GC and a weakened basal lamina, whereas GC migration was minimal in DBP-FW animals. Our findings provide the first evidence for how testicular dysgenesis can result after normal testis differentiation/development and may be relevant to understanding TDS in human patients.


Assuntos
Dibutilftalato/toxicidade , Disgenesia Gonadal/fisiopatologia , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Doenças Testiculares/fisiopatologia , Animais , Modelos Animais de Doenças , Feminino , Feto/efeitos dos fármacos , Feto/fisiopatologia , Disgenesia Gonadal/induzido quimicamente , Humanos , Células Intersticiais do Testículo/efeitos dos fármacos , Células Intersticiais do Testículo/patologia , Masculino , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Ratos , Ratos Wistar , Túbulos Seminíferos/efeitos dos fármacos , Túbulos Seminíferos/crescimento & desenvolvimento , Túbulos Seminíferos/patologia , Diferenciação Sexual/efeitos dos fármacos , Doenças Testiculares/induzido quimicamente , Testículo/efeitos dos fármacos , Testículo/crescimento & desenvolvimento , Testículo/patologia
4.
JCI Insight ; 2(6): e91204, 2017 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-28352662

RESUMO

The testicular dysgenesis syndrome (TDS) hypothesis, which proposes that common reproductive disorders of newborn and adult human males may have a common fetal origin, is largely untested. We tested this hypothesis using a rat model involving gestational exposure to dibutyl phthalate (DBP), which suppresses testosterone production by the fetal testis. We evaluated if induction of TDS via testosterone suppression is restricted to the "masculinization programming window" (MPW), as indicated by reduction in anogenital distance (AGD). We show that DBP suppresses fetal testosterone equally during and after the MPW, but only DBP exposure in the MPW causes reduced AGD, focal testicular dysgenesis, and TDS disorders (cryptorchidism, hypospadias, reduced adult testis size, and compensated adult Leydig cell failure). Focal testicular dysgenesis, reduced size of adult male reproductive organs, and TDS disorders and their severity were all strongly associated with reduced AGD. We related our findings to human TDS cases by demonstrating similar focal dysgenetic changes in testes of men with preinvasive germ cell neoplasia (GCNIS) and in testes of DBP-MPW animals. If our results are translatable to humans, they suggest that identification of potential causes of human TDS disorders should focus on exposures during a human MPW equivalent, especially if negatively associated with offspring AGD.


Assuntos
Disgenesia Gonadal/induzido quimicamente , Doenças Testiculares/induzido quimicamente , Animais , Dibutilftalato/toxicidade , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Exposição Materna , Plastificantes/toxicidade , Ratos
5.
Sci Rep ; 6: 19789, 2016 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-26813099

RESUMO

Analgesics which affect prostaglandin (PG) pathways are used by most pregnant women. As germ cells (GC) undergo developmental and epigenetic changes in fetal life and are PG targets, we investigated if exposure of pregnant rats to analgesics (indomethacin or acetaminophen) affected GC development and reproductive function in resulting offspring (F1) or in the F2 generation. Exposure to either analgesic reduced F1 fetal GC number in both sexes and altered the tempo of fetal GC development sex-dependently, with delayed meiotic entry in oogonia but accelerated GC differentiation in males. These effects persisted in adult F1 females as reduced ovarian and litter size, whereas F1 males recovered normal GC numbers and fertility by adulthood. F2 offspring deriving from an analgesic-exposed F1 parent also exhibited sex-specific changes. F2 males exhibited normal reproductive development whereas F2 females had smaller ovaries and reduced follicle numbers during puberty/adulthood; as similar changes were found for F2 offspring of analgesic-exposed F1 fathers or mothers, we interpret this as potentially indicating an analgesic-induced change to GC in F1. Assuming our results are translatable to humans, they raise concerns that analgesic use in pregnancy could potentially affect fertility of resulting daughters and grand-daughters.


Assuntos
Analgésicos/farmacologia , Células Germinativas/citologia , Células Germinativas/efeitos dos fármacos , Exposição Materna , Efeitos Tardios da Exposição Pré-Natal , Reprodução/efeitos dos fármacos , Animais , Diferenciação Celular , Feminino , Feto , Masculino , Fenótipo , Gravidez , Prostaglandinas/metabolismo , Ratos
6.
Environ Health Perspect ; 123(3): 223-30, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25514601

RESUMO

BACKGROUND: Phthalate exposure induces germ cell effects in the fetal rat testis. Although experimental models have shown that the human fetal testis is insensitive to the steroidogenic effects of phthalates, the effects on germ cells have been less explored. OBJECTIVES: We sought to identify the effects of phthalate exposure on human fetal germ cells in a dynamic model and to establish whether the rat is an appropriate model for investigating such effects. METHODS: We used immunohistochemistry, immunofluorescence, and quantitative real-time polymerase chain reaction to examine Sertoli and germ cell markers on rat testes and human fetal testis xenografts after exposure to vehicle or di(n-butyl) phthalate (DBP). Our study included analysis of germ cell differentiation markers, proliferation markers, and cell adhesion proteins. RESULTS: In both rat and human fetal testes, DBP exposure induced similar germ cell effects, namely, germ cell loss (predominantly undifferentiated), induction of multinucleated gonocytes (MNGs), and aggregation of differentiated germ cells, although the latter occurred rarely in the human testes. The mechanism for germ cell aggregation and MNG induction appears to be loss of Sertoli cell-germ cell membrane adhesion, probably due to Sertoli cell microfilament redistribution. CONCLUSIONS: Our findings provide the first comparison of DBP effects on germ cell number, differentiation, and aggregation in human testis xenografts and in vivo in rats. We observed comparable effects on germ cells in both species, but the effects in the human were muted compared with those in the rat. Nevertheless, phthalate effects on germ cells have potential implications for the next generation, which merits further study. Our results indicate that the rat is a human-relevant model in which to explore the mechanisms for germ cell effects.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Dibutilftalato/toxicidade , Células Germinativas/efeitos dos fármacos , Substâncias Perigosas/toxicidade , Testículo/efeitos dos fármacos , Animais , Feto/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Masculino , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Testículo/embriologia , Transplante Heterólogo
7.
Proc Natl Acad Sci U S A ; 111(18): E1924-32, 2014 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-24753613

RESUMO

Fetal growth plays a role in programming of adult cardiometabolic disorders, which in men, are associated with lowered testosterone levels. Fetal growth and fetal androgen exposure can also predetermine testosterone levels in men, although how is unknown, because the adult Leydig cells (ALCs) that produce testosterone do not differentiate until puberty. To explain this conundrum, we hypothesized that stem cells for ALCs must be present in the fetal testis and might be susceptible to programming by fetal androgen exposure during masculinization. To address this hypothesis, we used ALC ablation/regeneration to identify that, in rats, ALCs derive from stem/progenitor cells that express chicken ovalbumin upstream promoter transcription factor II. These stem cells are abundant in the fetal testis of humans and rodents, and lineage tracing in mice shows that they develop into ALCs. The stem cells also express androgen receptors (ARs). Reduction in fetal androgen action through AR KO in mice or dibutyl phthalate (DBP) -induced reduction in intratesticular testosterone in rats reduced ALC stem cell number by ∼40% at birth to adulthood and induced compensated ALC failure (low/normal testosterone and elevated luteinizing hormone). In DBP-exposed males, this failure was probably explained by reduced testicular steroidogenic acute regulatory protein expression, which is associated with increased histone methylation (H3K27me3) in the proximal promoter. Accordingly, ALCs and ALC stem cells immunoexpressed increased H3K27me3, a change that was also evident in ALC stem cells in fetal testes. These studies highlight how a key component of male reproductive development can fundamentally reprogram adult hormone production (through an epigenetic change), which might affect lifetime disease risk.


Assuntos
Células-Tronco Adultas/fisiologia , Androgênios/fisiologia , Desenvolvimento Fetal/fisiologia , Células Intersticiais do Testículo/fisiologia , Células-Tronco Adultas/efeitos dos fármacos , Animais , Callithrix , Linhagem da Célula/fisiologia , Dibutilftalato/toxicidade , Feminino , Desenvolvimento Fetal/efeitos dos fármacos , Células-Tronco Fetais/efeitos dos fármacos , Células-Tronco Fetais/fisiologia , Humanos , Técnicas In Vitro , Células Intersticiais do Testículo/efeitos dos fármacos , Hormônio Luteinizante/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Animais , Gravidez , Ratos , Ratos Transgênicos , Ratos Wistar , Receptores Androgênicos/deficiência , Receptores Androgênicos/genética , Receptores Androgênicos/fisiologia , Regeneração , Testículo/embriologia , Testículo/fisiologia , Testosterona/deficiência , Testosterona/fisiologia
8.
PLoS One ; 8(4): e61726, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23620786

RESUMO

In rodents, in utero exposure to exogenous estrogens including diethylstilboestrol (DES) results in major suppression of steroidogenesis in fetal testes. Whether similar effects occur in the human fetal testis is equivocal. Based on the results of the rodent studies, we hypothesised that exposure of human fetal testes to DES would result in a reduction in testosterone production. We show, using a xenograft approach, that testosterone production is not reduced in human fetal testis following DES exposure. Human fetal testes (15-19 weeks' gestation, n = 6) were xenografted into castrate male nude mice which were then treated for 35 days with vehicle or 100 µg/kg DES three times a week. For comparison, similar treatment was applied to pregnant rats from e13.5-e20.5 and effects on fetal testes evaluated at e21.5. Xenograft testosterone production was assessed by measuring host seminal vesicle (SV) weights as an indirect measure over the entire grafting period, and single measurement of serum testosterone at termination. Human fetal testis xenografts showed similar survival in DES and vehicle-exposed hosts. SV weight (44.3 v 26.6 mg, p = 0.01) was significantly increased in DES compared to vehicle-exposed hosts, respectively, indicating an overall increase in xenograft testosterone production over the grafting period, whilst serum testosterone at termination was unchanged. In contrast intra-testicular testosterone levels were reduced by 89%, in fetal rats exposed to DES. In rats, DES effects are mediated via Estrogen Receptor α (ESR1). We determined ESR1 protein and mRNA expression in human and rat fetal testis. ESR1 was expressed in rat, but not in human, fetal Leydig cells. We conclude that human fetal testis exposure to DES does not impair testosterone production as it does in rats, probably because ESR1 is not expressed in human fetal Leydig cells. This indicates that DES exposure is likely to pose minimal risk to masculinization of the human fetus.


Assuntos
Dietilestilbestrol/farmacologia , Feto/metabolismo , Testículo/embriologia , Testículo/transplante , Testosterona/biossíntese , Transplante Heterólogo , Animais , Dietilestilbestrol/administração & dosagem , Embrião de Mamíferos/efeitos dos fármacos , Endométrio/efeitos dos fármacos , Endométrio/metabolismo , Receptor alfa de Estrogênio/metabolismo , Feminino , Feto/efeitos dos fármacos , Sobrevivência de Enxerto/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Nus , Gravidez , Ratos , Ratos Wistar , Testículo/efeitos dos fármacos , Testículo/metabolismo
9.
Toxicol Sci ; 132(2): 443-57, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23358192

RESUMO

Previous analysis of in utero dibutylphthalate (DBP)-exposed fetal rat testes indicated that DBP's antiandrogenic effects were mediated, in part, by indirect inhibition of steroidogenic factor 1 (SF1), suggesting that peroxisome proliferator-activated receptor alpha (PPARα) might be involved through coactivator (CREB-binding protein [CBP]) sequestration. To test this hypothesis, we have performed chromatin immunoprecipitation (ChIP) microarray analysis to assess the DNA binding of PPARα, SF1, CBP, and RNA polymerase II in DBP-induced testicular maldevelopment target genes. Pathway analysis of expression array data in fetal rat testes examined at gestational day (GD) 15, 17, or 19 indicated that lipid metabolism genes regulated by SF1 and PPARα, respectively, were overrepresented, and the time dependency of changes to PPARα-regulated lipid metabolism genes correlated with DBP-mediated repression of SF1-regulated steroidogenesis genes. ChIP microarrays were used to investigate whether DBP-mediated repression of SF1-regulated genes was associated with changes in SF1 binding to genes involved in DBP-induced testicular maldevelopment. DBP treatment caused reductions in SF1 binding in CYP11a, StAR, and CYP17a. Follicle-stimulating hormone receptor (FSHR), regulated by SF1 but unaffected by DBP-treatment, also contained SF1-binding peaks, but DBP did not change this compared with control. GD15 and GD19 fetal testes contained PPARα protein-binding peaks in CYP11a, StAR, and CYP17a regulatory regions. In contrast to its repressive effect on SF1, DBP treatment caused increases in these peaks compared with control. PPARα-binding peaks in the FSHR promoter were not detected in GD15 samples. Hence, the repressive effect of DBP on SF1-regulated steroidogenic genes correlates with inhibition of SF1-DNA binding and increased PPARα-DNA binding. The data indicate that PPARα may act as an indirect transrepressor of SF1 on steroidogenic genes in fetal rat testes in response to DBP treatment.


Assuntos
Dibutilftalato/toxicidade , Testículo/efeitos dos fármacos , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Animais , Imunoprecipitação da Cromatina , Feminino , Imunoquímica , Masculino , Ratos , Ratos Wistar , Testículo/metabolismo
10.
J Clin Endocrinol Metab ; 96(11): E1746-55, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21865353

RESUMO

CONTEXT: The endometrium is a multicellular, steroid-responsive tissue that undergoes dynamic remodeling every menstrual cycle in preparation for implantation and, in absence of pregnancy, menstruation. Androgen receptors are present in the endometrium. OBJECTIVE: The objective of the study was to investigate the impact of androgens on human endometrial stromal cells (hESC). DESIGN: Bioinformatics was used to identify an androgen-regulated gene set and processes associated with their function. Regulation of target genes and impact of androgens on cell function were validated using primary hESC. SETTING: The study was conducted at the University Research Institute. PATIENTS: Endometrium was collected from women with regular menses; tissues were used for recovery of cells, total mRNA, or protein and for immunohistochemistry. RESULTS: A new endometrial androgen target gene set (n = 15) was identified. Bioinformatics revealed 12 of these genes interacted in one pathway and identified an association with control of cell survival. Dynamic androgen-dependent changes in expression of the gene set were detected in hESC with nine significantly down-regulated at 2 and/or 8 h. Treatment of hESC with dihydrotestosterone reduced staurosporine-induced apoptosis and cell migration/proliferation. CONCLUSIONS: Rigorous in silico analysis resulted in identification of a group of androgen-regulated genes expressed in human endometrium. Pathway analysis and functional assays suggest androgen-dependent changes in gene expression may have a significant impact on stromal cell proliferation, migration, and survival. These data provide the platform for further studies on the role of circulatory or local androgens in the regulation of endometrial function and identify androgens as candidates in the pathogenesis of common endometrial disorders including polycystic ovarian syndrome, cancer, and endometriosis.


Assuntos
Androgênios/farmacologia , Apoptose/efeitos dos fármacos , Endométrio/efeitos dos fármacos , Células Estromais/efeitos dos fármacos , Adulto , Androgênios/metabolismo , Apoptose/fisiologia , Endométrio/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Regiões Promotoras Genéticas , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Células Estromais/metabolismo
11.
J Clin Endocrinol Metab ; 95(10): E224-8, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20668045

RESUMO

CONTEXT: Differentiation (decidualization) of endometrial stromal cells (ESC) is an essential prerequisite for successful implantation and establishment of pregnancy. OBJECTIVE: The aim was to determine whether the orphan nuclear receptor estrogen-related receptor α (ERRα, NR3B1), and its target genes, medium chain specific acyl-CoA dehydrogenase (MCAD, ACADM), pyruvate dehydrogenase kinase 4 (PDK4), and phosphoenolpyruvate carboxykinase 2 (PEPCK, PCK2), play a role in the decidualization process. SETTING: We conducted the study at a University Research Institute. PATIENTS AND METHODS: Endometrial tissues were collected from women with regular menstrual cycles; tissues were used for recovery of primary ESC or RNA extraction or were fixed for immunohistochemistry. Primary ESC were decidualized in vitro; some cells were treated with XCT790 (ERRα inverse agonist). RESULTS: Decidualization of ESC in vitro was associated with a significant increase in expression of transcripts encoding ERRα and its coactivator peroxisome proliferator-activated receptor γ coactivator-1 α. Expression of ERRα target genes was altered with increased expression of MCAD and PDK4 and reduced expression of PEPCK. Incubation of decidualized ESC with XCT790 reduced expression of ERRα and markers of decidualization such as IGFBP-1. CONCLUSION: Increased expression of ERRα may play a role in altering the bioenergetics of decidualized ESC in preparation for implantation of the embryo and successful establishment of early pregnancy.


Assuntos
Decídua/fisiologia , Metabolismo Energético/genética , Regulação da Expressão Gênica , Receptores de Estrogênio/fisiologia , Células Estromais/fisiologia , Adulto , Técnicas de Cultura de Células , Células Cultivadas , Decídua/efeitos dos fármacos , Decídua/metabolismo , Implantação do Embrião/efeitos dos fármacos , Implantação do Embrião/genética , Endométrio/efeitos dos fármacos , Endométrio/metabolismo , Endométrio/fisiologia , Metabolismo Energético/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Antagonistas de Hormônios/farmacologia , Humanos , Nitrilas/farmacologia , Gravidez , Receptores de Estrogênio/antagonistas & inibidores , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Tiazóis/farmacologia , Receptor ERRalfa Relacionado ao Estrogênio
12.
BMC Cancer ; 9: 330, 2009 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-19758455

RESUMO

BACKGROUND: Endometrial cancer is the most common gynaecological malignancy; risk factors include exposure to oestrogens and high body mass index. Expression of enzymes involved in biosynthesis of oestrogens and prostaglandins (PG) is often higher in endometrial cancers when compared with levels detected in normal endometrium. Oestrogens bind one of two receptors (ERalpha and ERbeta) encoded by separate genes. The full-length receptors function as ligand-activated transcription factors; splice variant isoforms of ERbeta lacking a ligand-binding domain have also been described. PGs act in an autocrine or paracrine manner by binding to specific G-protein coupled receptors. METHODS: We compared expression of ERs, progesterone receptor (PR) and cyclooxygenase-2 (COX-2) in stage 1 endometrial adenocarcinomas graded as well (G1), moderately (G2) or poorly (G3) differentiated (n >or= 10 each group) using qRTPCR, single and double immunohistochemistry. We used endometrial adenocarcinoma cell lines to investigate the impact of PGF2alpha on expression of ERs and PR. RESULTS: Full length ERbeta (ERbeta1) and two ERbeta variants (ERbeta2, ERbeta5) were expressed in endometrial cancers regardless of grade and the proteins were immunolocalised to the nuclei of cells in both epithelial and stromal compartments. Immunoexpression of COX-2 was most intense in cells that were ERalphaneg/low. Expression of PR in endometrial adenocarcinoma (Ishikawa) cell lines and tissues broadly paralleled that of ERalpha. Treatment of adenocarcinoma cells with PGF2alpha reduced expression of ERalpha but had no impact on ERbeta1. Cells incubated with PGF2alpha were unable to increase expression of PR mRNA when they were incubated with E2. CONCLUSION: We have demonstrated that ERbeta5 protein is expressed in stage 1 endometrial adenocarcinomas. Expression of three ERbeta variants, including the full-length protein is not grade-dependent and most cells in poorly differentiated cancers are ERbetapos/ERalphaneg. We found evidence of a link between COX-2, its product PGF2alpha, and expression of ERalpha and PR that sheds new light on the cross talk between steroid and PG signalling pathways in this disease.


Assuntos
Dinoprosta/metabolismo , Neoplasias do Endométrio/genética , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/genética , Regulação Neoplásica da Expressão Gênica , Adulto , Linhagem Celular Tumoral , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Dinoprosta/genética , Neoplasias do Endométrio/metabolismo , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Feminino , Variação Genética , Humanos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo
13.
Hum Reprod ; 23(12): 2782-90, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18775884

RESUMO

BACKGROUND: Estrogen receptor related beta (ERRbeta, ESRRB/NR3B2) is an orphan receptor that shares significant sequence homology with estrogen receptors ERalpha and ERbeta. ERR family members are reported to exhibit constitutive transcriptional activity; however, little is known about the biological function of ERRbeta. In an attempt to delineate its role, we examined expression of ERRbeta in normal human endometrium, a tissue that undergoes cyclic remodelling under the influence of estrogen and progesterone. METHODS: Well-characterized endometrial tissue (n = 31), including full-thickness biopsies, was obtained from women with regular menstrual cycles. RT-PCR was used to measure mRNA encoding ERRbeta, the peroxisome proliferator activated receptor gamma coactivators (PGC)-1alpha and beta and to determine whether ERRbeta splice variant mRNAs were expressed. ERRbeta was immunolocalized using both single and double antibody immunohistochemistry. RESULTS: Total ERRbeta mRNA appeared higher in proliferative phase samples but results did not reach significance. Transcripts corresponding to the long- and short-splice variants of ERRbeta as well as PGC1alpha and beta were detected but ERRbetaDelta10 was absent. ERRbeta protein was localized to cell nuclei within multiple endometrial cell types including the glands, stroma, endothelium and immune cells, including uterine natural killer (uNK) cells and macrophages. Fluorescent immunohistochemistry revealed that some cells co-expressed ERRbeta and ERalpha or ERbeta, for example, endothelial and uNK cells were ERRbeta+/ERbeta+. CONCLUSIONS: ERRbeta mRNA and protein are expressed in healthy human endometrium. Further studies are warranted to characterize the functional impact of ERRbeta on endometrial biology.


Assuntos
Endométrio/metabolismo , Ciclo Menstrual/fisiologia , Receptores de Estrogênio/biossíntese , Adulto , Proteínas de Transporte/biossíntese , Endométrio/citologia , Receptor alfa de Estrogênio/biossíntese , Receptor beta de Estrogênio/biossíntese , Feminino , Expressão Gênica , Proteínas de Choque Térmico/biossíntese , Humanos , Células Matadoras Naturais/metabolismo , Leucócitos/metabolismo , Pessoa de Meia-Idade , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Isoformas de Proteínas/biossíntese , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA , Fatores de Transcrição/biossíntese
14.
Steroids ; 67(12): 985-92, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12398995

RESUMO

Two structurally related subtypes of oestrogen receptor (ER), known as alpha (ER alpha, NR3A1) and beta (ER beta, NR3A2) have been identified. ER beta mRNA and protein have been detected in a wide range of tissues including the vasculature, bone, and gonads in both males and females, as well as in cancers of the breast and prostate. In many tissues the pattern of expression of ER beta is distinct from that of ER alpha. A number of variant isoforms of the wild type beta receptor (ER beta 1), have been identified. In the human these include: (1). use of alternative start sites within the mRNA leading to translation of either a long (530 amino acids, hER beta 1L) or a truncated form (487aa hER beta 1s); (2). deletion of exons by alternative splicing; (3). formation of several isoforms (ER beta 2-beta 5) due to alternative splicing of exons encoding the carboxy terminus (F domain). We have raised monoclonal antibodies specific for hER beta1 as well as to three of the C terminal isoforms (beta2, beta 4 and beta 5). Using these antibodies we have found that ER beta 2, beta 4 and beta 5 proteins are expressed in nuclei of human tissues including the ovary, placenta, testis and vas deferens. In conclusion, in addition to the differential expression of full length ER alpha and ER beta a number of ER variant isoforms have been identified. The impact of the expression of these isoforms on cell responsiveness to oestrogens may add additional complexity to the ways in which oestrogenic ligands influence cell function.


Assuntos
Receptores de Estrogênio/biossíntese , Processamento Alternativo , Animais , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Feminino , Humanos , Masculino , Camundongos , Placenta/metabolismo , Isoformas de Proteínas/genética , RNA Mensageiro/metabolismo , Receptores de Estrogênio/genética , Útero/metabolismo , Ducto Deferente/metabolismo
15.
J Clin Endocrinol Metab ; 87(6): 2706-15, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12050238

RESUMO

Estrogens can regulate germ cell function. Estrogen action is mediated via high affinity ERs; two subtypes (ERalpha and ERbeta) have been identified. We have shown previously that ERbeta is expressed in nuclei of multiple human testicular cells. A variant isoform of human (h) ERbeta (hERbetacx/2), formed by alternative splicing, has been identified in testicular cDNA libraries by two laboratories. The present study examined the expression of wild-type (ERbeta1) and variant (ERbeta2) beta receptors in human testes by 1) RT-PCR with isoform specific primers, and 2) single and double immunohistochemistry using monoclonal antibodies raised against peptides unique to the C termini of hERbeta1 and hERbeta2. PCR products specific for ERbeta1 and ERbeta2 were amplified from cDNA pools prepared from human testes and granulosa cells. On Western blots, the anti-ERbeta1 monoclonal antibody bound to recombinant ERbeta1 and the anti-ERbeta2 monoclonal to recombinant hERbeta2. Neither bound to the other ERbeta isoform nor to recombinant ERalpha. ERbeta1 and ERbeta2 proteins were both detected in human testis. Immunoexpression of ERbeta1 was most intense in pachytene spermatocytes and round spermatids, whereas low levels of expression were detected in Sertoli cells, spermatogonia, preleptotene, leptotene, zygotene, and diplotene spermatocytes. Highest levels of expression of ERbeta2 protein were detected in Sertoli cells and spermatogonia with low/variable expression in preleptotene, pachytene, and diplotene spermatocytes. No immunostaining was detected in elongating spermatids. Most interstitial cells expressed more ERbeta2 than ERbeta1. It is speculated that the cells most susceptible to modulation by estrogenic ligands are round spermatids in which levels of expression of ERbeta1 are high. In contrast, expression of ERbeta2, an isoform that may act as a dominant negative inhibitor of ER action, in Sertoli cells and spermatogonia, could protect these cells from adverse effects of estrogens.


Assuntos
DNA Recombinante , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Testículo/metabolismo , Adulto , Anticorpos Monoclonais , Receptor beta de Estrogênio , Imunofluorescência , Variação Genética , Humanos , Imuno-Histoquímica , Masculino , Isoformas de Proteínas/metabolismo , Testículo/citologia , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA