Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
BMC Microbiol ; 19(1): 57, 2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30871463

RESUMEN

BACKGROUND: A majority of women with polycystic ovary syndrome (PCOS) have metabolic dysfunction that results in an increased risk of type 2 diabetes. We previously developed a pubertal mouse model using the aromatase inhibitor, letrozole, which recapitulates many of the reproductive and metabolic features of PCOS. To further our understanding of the effects of androgen excess, we compared the effects of letrozole treatment initiated in puberty versus adulthood on reproductive and metabolic phenotypes as well as on the gut microbiome. RESULTS: Letrozole treatment of both pubertal and adult female mice resulted in reproductive hallmarks of PCOS, including hyperandrogenemia, anovulation and polycystic ovaries. However, unlike pubertal mice, treatment of adult female mice resulted in modest weight gain and abdominal adiposity, minimal elevation in fasting blood glucose and insulin levels, and no detectable insulin resistance. In addition, letrozole treatment of adult mice was associated with a distinct shift in gut microbial diversity compared to letrozole treatment of pubertal mice. CONCLUSIONS: Our results indicate that dysregulation of metabolism and the gut microbiome in PCOS may be influenced by the timing of androgen exposure. In addition, the minimal weight gain and lack of insulin resistance in adult female mice after letrozole treatment indicates that this model may be useful for investigating the effects of hyperandrogenemia on the hypothalamic-pituitary-gonadal axis and the periphery without the influence of substantial metabolic dysregulation.


Asunto(s)
Inhibidores de la Aromatasa/administración & dosificación , Microbioma Gastrointestinal/efectos de los fármacos , Letrozol/administración & dosificación , Síndrome del Ovario Poliquístico/metabolismo , Síndrome del Ovario Poliquístico/microbiología , Factores de Edad , Animales , Modelos Animales de Enfermedad , Femenino , Resistencia a la Insulina , Ratones , Ratones Endogámicos C57BL , Fenotipo , Síndrome del Ovario Poliquístico/inducido químicamente , Aumento de Peso
2.
J Biol Chem ; 287(40): 33424-35, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-22865884

RESUMEN

Synthesis of luteinizing hormone (LH) is tightly controlled by a complex network of hormonal signaling pathways that can be modulated by metabolic cues, such as insulin. One group of candidate genes that may be regulated by insulin signaling in pituitary gonadotrope cells is the FOXO subfamily of forkhead transcription factors. In this study we investigated whether FOXO1 is expressed in gonadotropes and if it can modulate LH ß-subunit (Lhb) gene expression. We demonstrated that FOXO1 is expressed in murine gonadotrope cells and that insulin signaling increased FOXO1 phosphorylation and cytoplasmic localization in a PI3K-dependent manner. We also showed that FOXO1 repressed basal transcription and gonadotropin-releasing hormone (GnRH) induction of both the murine and human LHB genes in LßT2 cells, suggesting that FOXO1 regulation of LHB transcription may be conserved between rodents and humans. Although we did not detect FOXO1 binding to the proximal Lhb promoter, the FOXO1 DNA binding domain was necessary for the suppression, suggesting that FOXO1 exerts its effect through protein-protein interactions with transcription factors/cofactors required for Lhb gene expression. FOXO1 repression mapped to the proximal Lhb promoter containing steroidogenic factor 1 (SF1), pituitary homeobox 1 (PTX1), and early growth response protein 1 (EGR1) binding elements. Additionally, FOXO1 blocked induction of the Lhb promoter with overexpressed SF1, PTX1, and EGR1, indicating that FOXO1 repression occurs via these transcription factors but not through regulation of their promoters. In summary, we demonstrate that FOXO1 phosphorylation and cellular localization is regulated by insulin signaling in gonadotropes and that FOXO1 inhibits Lhb transcription. Our study also suggests that FOXO1 may play an important role in controlling LH levels in response to metabolic cues.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Hormona Luteinizante de Subunidad beta/metabolismo , Animales , Línea Celular , Núcleo Celular/metabolismo , Femenino , Proteína Forkhead Box O1 , Humanos , Inmunohistoquímica/métodos , Insulina/metabolismo , Hormona Luteinizante/metabolismo , Masculino , Ratones , Fosforilación , Hipófisis/metabolismo , Transducción de Señal , Transcripción Genética
3.
Biochim Biophys Acta ; 1782(4): 259-70, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18280813

RESUMEN

Ser/Thr protein phosphatase 5 (PP5) regulates several signaling-cascades that suppress growth and/or facilitate apoptosis in response to genomic stress. The expression of PP5 is responsive to hypoxia inducible factor-1 (HIF-1) and estrogen, which have both been linked to the progression of human breast cancer. Still, it is not clear if PP5 plays a role in the development of human cancer. Here, immunostaining of breast cancer tissue-microarrays (TMAs) revealed a positive correlation between PP5 over-expression and ductal carcinoma in situ (DCIS; P value 0.0028), invasive ductal carcinoma (IDC; P value 0.012) and IDC with metastases at the time of diagnosis (P value 0.0001). In a mouse xenograft model, the constitutive over-expression of PP5 was associated with an increase in the rate of tumor growth. In a MCF-7 cell culture model over-expression correlated with both an increase in the rate of proliferation and protection from cell death induced by oxidative stress, UVC-irradiation, adriamycin, and vinblastine. PP5 over-expression had no apparent effect on the sensitivity of MCF-7 cells to taxol or rapamycin. Western analysis of extracts from cells over-expressing PP5 revealed a decrease in the phosphorylation of known substrates for PP5. Together, these studies indicate that elevated levels of PP5 protein occur in human breast cancer and suggest that PP5 over-expression may aid tumor progression.


Asunto(s)
Neoplasias de la Mama/enzimología , Carcinoma Ductal de Mama/enzimología , Carcinoma Intraductal no Infiltrante/enzimología , Animales , Muerte Celular , Línea Celular Tumoral , Doxorrubicina/farmacología , Humanos , Ratones , Ratones Desnudos , Proteínas Nucleares , Estrés Oxidativo , Fosfoproteínas Fosfatasas , Treonina , Factores de Tiempo , Rayos Ultravioleta , Regulación hacia Arriba , Vinblastina/farmacología
4.
Endocrinology ; 158(9): 2988-3003, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28911175

RESUMEN

Women with polycystic ovary syndrome (PCOS) diagnosed with hyperandrogenism and ovulatory dysfunction have an increased risk of developing metabolic disorders, including type 2 diabetes and cardiovascular disease. We previously developed a model that uses letrozole to elevate endogenous testosterone levels in female mice. This model has hallmarks of PCOS, including hyperandrogenism, anovulation, and polycystic ovaries, as well as increased abdominal adiposity and glucose intolerance. In the current study, we further characterized the metabolic dysfunction that occurs after letrozole treatment to determine whether this model represents a PCOS-like metabolic phenotype. We focused on whether letrozole treatment results in altered pancreatic or liver function as well as insulin resistance. We also investigated whether hyperinsulinemia occurs secondary to weight gain and insulin resistance in this model or if it can occur independently. Our study demonstrated that letrozole-treated mice developed hyperinsulinemia after 1 week of treatment and without evidence of insulin resistance. After 2 weeks of letrozole treatment, mice became significantly heavier than placebo mice, demonstrating that weight gain was not required to develop hyperinsulinemia. After 5 weeks of letrozole treatment, mice exhibited blunted glucose-stimulated insulin secretion, insulin resistance, and impaired insulin-induced phosphorylation of AKT in skeletal muscle. Moreover, letrozole-treated mice exhibited dyslipidemia after 5 weeks of treatment but no evidence of hepatic disease. Our study demonstrated that the letrozole-induced PCOS mouse model exhibits multiple features of the metabolic dysregulation observed in obese, hyperandrogenic women with PCOS. This model will be useful for mechanistic studies investigating how hyperandrogenemia affects metabolism in females.


Asunto(s)
Hiperandrogenismo/inducido químicamente , Hiperandrogenismo/complicaciones , Hiperinsulinismo/etiología , Resistencia a la Insulina , Nitrilos/farmacología , Maduración Sexual/efectos de los fármacos , Triazoles/farmacología , Aumento de Peso/efectos de los fármacos , Animales , Ingestión de Alimentos/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Femenino , Glucosa/metabolismo , Hiperinsulinismo/metabolismo , Letrozol , Ratones , Ratones Endogámicos C57BL , Obesidad/inducido químicamente , Obesidad/metabolismo
5.
PLoS One ; 11(1): e0146509, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26731268

RESUMEN

Women with polycystic ovary syndrome (PCOS) have reproductive and metabolic abnormalities that result in an increased risk of infertility, diabetes and cardiovascular disease. The large intestine contains a complex community of microorganisms (the gut microbiome) that is dysregulated in humans with obesity and type 2 diabetes. Using a letrozole-induced PCOS mouse model, we demonstrated significant diet-independent changes in the gut microbial community, suggesting that gut microbiome dysbiosis may also occur in PCOS women. Letrozole treatment was associated with a time-dependent shift in the gut microbiome and a substantial reduction in overall species and phylogenetic richness. Letrozole treatment also correlated with significant changes in the abundance of specific Bacteroidetes and Firmicutes previously implicated in other mouse models of metabolic disease in a time-dependent manner. Our results suggest that the hyperandrogenemia observed in PCOS may significantly alter the gut microbiome independently of diet.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Síndrome del Ovario Poliquístico/microbiología , Animales , Dieta , Modelos Animales de Enfermedad , Femenino , Letrozol , Ratones , Nitrilos , Síndrome del Ovario Poliquístico/inducido químicamente , Triazoles
6.
Mol Cell Endocrinol ; 405: 14-24, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25676570

RESUMEN

The FOXO1 transcription factor is important for multiple aspects of reproductive function. We previously reported that FOXO1 functions as a repressor of gonadotropin hormone synthesis, but how FOXO1 is regulated in pituitary gonadotropes is unknown. The growth factors, insulin and insulin-like growth factor I (IGF1), function as key regulators of cell proliferation, metabolism and apoptosis in multiple cell types through the PI3K/AKT signaling pathway. In this study, we found that insulin and IGF1 signaling in gonadotropes induced FOXO1 phosphorylation through the PI3K/AKT pathway in immortalized and primary cells, resulting in FOXO1 relocation from the nucleus to the cytoplasm. Furthermore, insulin administration in vivo induced phosphorylation of FOXO1 and AKT in the pituitary. Thus, insulin and IGF1 act as negative regulators of FOXO1 activity and may serve to fine-tune gonadotropin expression.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Gonadotrofos/metabolismo , Factor I del Crecimiento Similar a la Insulina/fisiología , Insulina/fisiología , Animales , Línea Celular , Proteína Forkhead Box O1 , Hormona Liberadora de Gonadotropina/fisiología , Ratones , Fosforilación , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-akt/metabolismo
7.
PLoS One ; 9(11): e113839, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25423188

RESUMEN

In the present study, we investigate whether the FOXO1 transcription factor modulates activin signaling in pituitary gonadotropes. Our studies show that overexpression of constitutively active FOXO1 decreases activin induction of murine Fshb gene expression in immortalized LßT2 cells. We demonstrate that FOXO1 suppression of activin induction maps to the -304/-95 region of the Fshb promoter containing multiple activin response elements and that the suppression requires the FOXO1 DNA-binding domain (DBD). FOXO1 binds weakly to the -125/-91 region of the Fshb promoter in a gel-shift assay. Since this region of the promoter contains a composite SMAD/FOXL2 binding element necessary for activin induction of Fshb transcription, it is possible that FOXO1 DNA binding interferes with SMAD and/or FOXL2 function. In addition, our studies demonstrate that FOXO1 directly interacts with SMAD3/4 but not SMAD2 in a FOXO1 DBD-dependent manner. Moreover, we show that SMAD3/4 induction of Fshb-luc and activin induction of a multimerized SMAD-binding element-luc are suppressed by FOXO1 in a DBD-dependent manner. These results suggest that FOXO1 binding to the proximal Fshb promoter as well as FOXO1 interaction with SMAD3/4 proteins may result in decreased activin induction of Fshb in gonadotropes.


Asunto(s)
Activinas/fisiología , Hormona Folículo Estimulante/genética , Factores de Transcripción Forkhead/fisiología , Gonadotrofos/metabolismo , Transcripción Genética/fisiología , Animales , Línea Celular Transformada , Proteína Forkhead Box O1 , Gonadotrofos/citología , Ratones , Regiones Promotoras Genéticas , ARN Mensajero/genética , Proteínas Smad/antagonistas & inhibidores , Proteínas Smad/fisiología
8.
Mol Endocrinol ; 27(11): 1825-39, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24065703

RESUMEN

Synthesis of the gonadotropin ß-subunits is tightly controlled by a complex network of hormonal signaling pathways that may be modulated by metabolic cues. Recently, we reported that insulin regulates FOXO1 phosphorylation and cellular localization in pituitary gonadotropes and that FOXO1 overexpression inhibits Lhb transcription. In the current study, we investigated whether FOXO1 modulates Fshb synthesis. Here, we demonstrate that FOXO1 represses basal and GnRH-induced Fshb transcription in LßT2 cells. In addition, we show that PI3K inhibition, which increases FOXO1 nuclear localization, results in decreased Fshb mRNA levels in murine primary pituitary cells. FOXO1 also decreases transcription from the human FSHB promoter, suggesting that FOXO1 regulation of FSHB transcription may be conserved between rodents and humans. Although the FOXO1 DNA-binding domain is necessary for suppression of Fshb, we do not observe direct binding of FOXO1 to the Fshb promoter, suggesting that FOXO1 exerts its effect through protein-protein interactions with transcription factors required for Fshb synthesis. FOXO1 suppression of basal Fshb transcription may involve PITX1 because PITX1 interacts with FOXO1, FOXO1 repression maps to the proximal Fshb promoter containing a PITX1-binding site, PITX1 induction of Fshb or a PITX1 binding element in CV-1 cells is decreased by FOXO1, and FOXO1 suppresses Pitx1 mRNA and protein levels. GnRH induction of an Fshb promoter containing a deletion at -50/-41 or -30/-21 is not repressed by FOXO1, suggesting that these two regions may be involved in FOXO1 suppression of GnRH-induced Fshb synthesis. In summary, our data demonstrate that FOXO1 can negatively regulate Fshb transcription and suggest that FOXO1 may relay metabolic hormonal signals to modulate gonadotropin production.


Asunto(s)
Hormona Folículo Estimulante de Subunidad beta/genética , Factores de Transcripción Forkhead/fisiología , Silenciador del Gen , Gonadotrofos/metabolismo , Hormona Liberadora de Gonadotropina/fisiología , Animales , Secuencia de Bases , Línea Celular , Hormona Folículo Estimulante de Subunidad beta/metabolismo , Proteína Forkhead Box O1 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA