Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Genet ; 17(3): e1009483, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33784295

RESUMEN

Conventionally viewed as male hormone, androgens play a critical role in female fertility. Although androgen receptors (AR) are transcription factors, to date very few direct transcriptional targets of ARs have been identified in the ovary. Using mouse models, this study provides three critical insights about androgen-induced gene regulation in the ovary and its impact on female fertility. First, RNA-sequencing reveals a number of genes and biological processes that were previously not known to be directly regulated by androgens in the ovary. Second, androgens can also influence gene expression by decreasing the tri-methyl mark on lysine 27 of histone3 (H3K27me3), a gene silencing epigenetic mark. ChIP-seq analyses highlight that androgen-induced modulation of H3K27me3 mark within gene bodies, promoters or distal enhancers have a much broader impact on ovarian function than the direct genomic effects of androgens. Third, androgen-induced decrease of H3K27me3 is mediated through (a) inhibiting the expression and activity of Enhancer of Zeste Homologue 2 (EZH2), a histone methyltransferase that promotes tri-methylation of K27 and (b) by inducing the expression of a histone demethylase called Jumonji domain containing protein-3 (JMJD3/KDM6B), responsible for removing the H3K27me3 mark. Androgens through the PI3K/Akt pathway, in a transcription-independent fashion, increase hypoxia-inducible factor 1 alpha (HIF1α) protein levels, which in turn induce JMJD3 expression. Furthermore, proof of concept studies involving in vivo knockdown of Ar in the ovary and ovarian (granulosa) cell-specific Ar knockout mouse model show that ARs regulate the expression of key ovarian genes through modulation of H3K27me3.


Asunto(s)
Andrógenos/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Regulación de la Expresión Génica , Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Ovario/metabolismo , Andrógenos/farmacología , Animales , Biología Computacional/métodos , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/metabolismo , Ratones , Ovario/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-akt , Transcriptoma
2.
Biol Reprod ; 107(3): 813-822, 2022 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-35657015

RESUMEN

The anti-Müllerian hormone (AMH) produced by the granulosa cells of growing follicles is critical for folliculogenesis and is clinically used as a diagnostic and prognostic marker of female fertility. Previous studies report that AMH-pretreatment in mice creates a pool of quiescent follicles that are released following superovulation, resulting in an increased number of ovulated oocytes. However, the quality and developmental competency of oocytes derived from AMH-induced accumulated follicles as well as the effect of AMH treatment on live birth are not known. This study reports that AMH priming positively affects oocyte maturation and early embryonic development culminating in higher number of live births. Our results show that AMH treatment results in good-quality oocytes with greater developmental competence that enhances embryonic development resulting in blastocysts with higher gene expression. The transcriptomic analysis of oocytes from AMH-primed mice compared with those of control mice reveal that AMH upregulates a large number of genes and pathways associated with oocyte quality and embryonic development. Mitochondrial function is the most affected pathway by AMH priming, which is supported by more abundant active mitochondria, mitochondrial DNA content and adenosine triphosphate levels in oocytes and embryos isolated from AMH-primed animals compared with control animals. These studies for the first time provide an insight into the overall impact of AMH on female fertility and highlight the critical knowledge necessary to develop AMH as a therapeutic option to improve female fertility.


Asunto(s)
Hormona Antimülleriana , Tasa de Natalidad , Animales , Hormona Antimülleriana/genética , Hormona Antimülleriana/metabolismo , Desarrollo Embrionario , Femenino , Nacimiento Vivo , Ratones , Oocitos/metabolismo , Folículo Ovárico/metabolismo , Embarazo
3.
J Dairy Sci ; 105(1): 842-855, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34696909

RESUMEN

Intense and protracted adipose tissue (AT) fat mobilization increases the risk of metabolic and inflammatory periparturient diseases in dairy cows. This vulnerability increases when cows have endotoxemia-common during periparturient diseases such as mastitis, metritis, and pneumonia-but the mechanisms are unknown. Fat mobilization intensity is determined by the balance between lipolysis and lipogenesis. Around parturition, the rate of lipolysis surpasses that of lipogenesis, leading to enhanced free fatty acid release into the circulation. We hypothesized that exposure to endotoxin (ET) increases AT lipolysis by activation of classic and inflammatory lipolytic pathways and reduction of insulin sensitivity. In experiment 1, subcutaneous AT (SCAT) explants were collected from periparturient (n = 12) Holstein cows at 11 ± 3.6 d (mean ± SE) before calving, and 6 ± 1 d and 13 ± 1.4 d after parturition. Explants were treated with the endotoxin lipopolysaccharide (LPS; 20 µg/mL; basal = 0 µg/mL) for 3 h. The effect of LPS on lipolysis was assessed in the presence of the ß-adrenergic agonist and promoter of lipolysis isoproterenol (ISO; 1 µM; LPS+ISO). In experiment 2, SCAT explants were harvested from 24 nonlactating, nongestating multiparous Holstein dairy cows and exposed to the same treatments as in experiment 1 for 3 and 7 h. The effect of LPS on the antilipolytic responses induced by insulin (INS = 1 µL/L, LPS+INS) was established during ISO stimulation [ISO+INS, LPS+ISO+INS]. The characterization of lipolysis included the quantification of glycerol release and the assessment of markers of lipase activity [adipose triglyceride lipase (ATGL), hormone-sensitive lipase (HSL), and phosphorylated HSL Ser563 (pHSL)], and insulin pathway activation (AKT, pAKT) using capillary electrophoresis. Inflammatory gene networks were evaluated by real-time quantitative PCR. In periparturient cows, LPS increased AT lipolysis by 67 ± 12% at 3 h across all time points compared with basal. In nonlactating cows, LPS was an effective lipolytic agent at 3 h and 7 h, increasing glycerol release by 115 ± 18% and 68.7 ± 16%, respectively, relative to basal. In experiment 2, LPS enhanced ATGL activity with minimal HSL activation at 3 h. In contrast, at 7 h, LPS increased HSL phosphorylation (i.e., HSL activity) by 123 ± 11%. The LPS-induced HSL lipolytic activity at 7 h coincided with the activation of the MEK/ERK inflammatory pathway. In experiment 2, INS reduced the lipolytic effect of ISO (ISO+INS: -63 ± 18%) and LPS (LPS+INS: -45.2 ± 18%) at 3 h. However, the antilipolytic effect of INS was lost in the presence of LPS at 7 h (LPS+INS: -16.3 ± 16%) and LPS+ISO+INS at 3 and 7 h (-3.84 ± 23.6% and -21.2 ± 14.6%). Accordingly, LPS reduced pAKT:AKT (0.11 ± 0.07) compared with basal (0.18 ± 0.05) at 7 h. Our results indicated that exposure to LPS activated the classic and inflammatory lipolytic pathways and reduced insulin sensitivity in SCAT. These data provide evidence that during endotoxemia, dairy cows may be more susceptible to lipolysis dysregulation and loss of adipocyte sensitivity to the antilipolytic action of insulin.


Asunto(s)
Enfermedades de los Bovinos , Resistencia a la Insulina , Tejido Adiposo/metabolismo , Animales , Bovinos , Enfermedades de los Bovinos/metabolismo , Femenino , Lipólisis , Lipopolisacáridos/metabolismo , Esterol Esterasa/metabolismo
4.
Biol Reprod ; 102(5): 1045-1054, 2020 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-31930385

RESUMEN

Maternal perturbations or sub-optimal conditions during fetal development can predispose the offspring to diseases in adult life. Animal and human studies show that prenatal androgen excess may be an underlying cause of polycystic ovary syndrome (PCOS) later in life. In women, PCOS is a common fertility disorder with comorbid metabolic dysfunction. Here, using a sheep model of PCOS phenotype, we elucidate the epigenetic changes induced by prenatal (30-90 day) testosterone (T) treatment and its effect on gene expression in fetal day 90 (D90) and adult year 2 (Y2) ovaries. RNA-seq study shows 65 and 99 differentially regulated genes in prenatal T-treated fetal and adult ovaries, respectively. Interestingly, there were no differences in gene inducing histone marks H3K27ac, H3K9ac, and H3K4me3 or in gene silencing marks, H3K27me3 and H3K9me3 in the fetal D90 ovaries of control and excess T-exposed fetuses. In contrast, except for H3K4me3 and H3K27me3, all the other histone marks were upregulated in the prenatal T-treated adult Y2 ovary. Chromatin immunoprecipitation (ChIP) studies in adult Y2 ovaries established a direct relationship between the epigenetic modifications with the upregulated and downregulated genes obtained from RNA-seq. Results show increased gene inducing marks, H3K27ac and H3K9ac, on the promoter region of upregulated genes while gene silencing mark, H3K9me3, was also significantly increased on the downregulated genes. This study provides a mechanistic insight into prenatal T-induced developmental programming and its effect on ovarian gene expression that may contribute to reproductive dysfunction and development of PCOS in adult life.


Asunto(s)
Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Ovario/efectos de los fármacos , Ovario/fisiología , Ovinos/fisiología , Testosterona/farmacología , Animales , Femenino , Desarrollo Fetal/efectos de los fármacos , Embarazo , Efectos Tardíos de la Exposición Prenatal , Ovinos/embriología
5.
Proc Natl Acad Sci U S A ; 111(8): 3008-13, 2014 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-24516121

RESUMEN

Although androgen excess is considered detrimental to women's health and fertility, global and ovarian granulosa cell-specific androgen-receptor (AR) knockout mouse models have been used to show that androgen actions through ARs are actually necessary for normal ovarian function and female fertility. Here we describe two AR-mediated pathways in granulosa cells that regulate ovarian follicular development and therefore female fertility. First, we show that androgens attenuate follicular atresia through nuclear and extranuclear signaling pathways by enhancing expression of the microRNA (miR) miR-125b, which in turn suppresses proapoptotic protein expression. Second, we demonstrate that, independent of transcription, androgens enhance follicle-stimulating hormone (FSH) receptor expression, which then augments FSH-mediated follicle growth and development. Interestingly, we find that the scaffold molecule paxillin regulates both processes, making it a critical regulator of AR actions in the ovary. Finally, we report that low doses of exogenous androgens enhance gonadotropin-induced ovulation in mice, further demonstrating the critical role that androgens play in follicular development and fertility. These data may explain reported positive effects of androgens on ovulation rates in women with diminished ovarian reserve. Furthermore, this study demonstrates mechanisms that might contribute to the unregulated follicle growth seen in diseases of excess androgens such as polycystic ovary syndrome.


Asunto(s)
Regulación de la Expresión Génica/fisiología , MicroARNs/metabolismo , Folículo Ovárico/crecimiento & desarrollo , Receptores de HFE/metabolismo , Testosterona/metabolismo , Análisis de Varianza , Animales , Western Blotting , Inmunoprecipitación de Cromatina , Femenino , Citometría de Flujo , Atresia Folicular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Células de la Granulosa/metabolismo , Etiquetado Corte-Fin in Situ , Ratones , Ratones Endogámicos C57BL , MicroARNs/antagonistas & inhibidores , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/metabolismo , Paxillin/genética , Paxillin/metabolismo , Testosterona/farmacología
6.
J Assist Reprod Genet ; 34(9): 1105-1113, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28643088

RESUMEN

Members of the transforming growth factor-beta (TGF-beta) superfamily are key regulators of various physiological processes. Anti-Müllerian hormone (AMH) which is also commonly known as Müllerian-inhibiting substance (MIS) is a member of the TGF-beta superfamily and an important regulator of reproductive organ differentiation and ovarian follicular development. While AMH has been used for diagnostic purposes as a biomarker for over 15 years, new potential therapeutic applications of recombinant human AMH analogues are now emerging as pharmacologic agents in reproductive medicine. Therapeutic uses of AMH in gonadal tissue may provide a unique opportunity to address a broad range of reproductive themes, like contraception, ovulation induction, onset of menopause, and fertility preservation, as well as specific disease conditions, such as polycystic ovarian syndrome (PCOS) and cancers of the reproductive tract. This review explores the most promising therapeutic applications for a novel class of drugs known as AMH analogues with agonist and antagonist functions.


Asunto(s)
Hormona Antimülleriana/uso terapéutico , Folículo Ovárico/efectos de los fármacos , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Medicina Reproductiva/tendencias , Hormona Antimülleriana/química , Femenino , Humanos , Inducción de la Ovulación/métodos , Síndrome del Ovario Poliquístico/patología , Reproducción/efectos de los fármacos
7.
J Transl Med ; 14(1): 172, 2016 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-27286817

RESUMEN

BACKGROUND: Though outcome models have been proposed previously, it is unknown whether cutoffs in clinical pregnancy and live birth rates at all ages are able to classify in vitro fertilization (IVF) patients into good-, intermediate- and poor prognosis. METHODS: We here in 3 infertile patient cohorts, involving 1247, 1514 and 632 women, built logistic regression models based on 3 functional ovarian reserve (FOR) parameters, including (1) number of good quality embryos, (2) follicle stimulating hormone (FSH, mIU/mL) and (3) anti-Müllerian hormone (AMH, ng/mL), determining whether clinical pregnancy and live birth rates can discriminate between good, intermediate and poor prognosis patients. RESULTS: All models, indeed, allowed at all ages for separation by prognosis, though cut offs changed with age. In the embryo model, increasing embryo production resulted in linear improvement of IVF outcomes despite transfer of similar embryo numbers; in the FSH model outcomes and FSH levels related inversely, while the association of AMH followed a bell-shaped polynomial pattern, demonstrating "best" outcomes at mid-ranges. All 3 models demonstrated increasingly poor outcomes with advancing ages, though "best" AMH even above age 43 was still associated with unexpectedly good pregnancy and delivery outcomes. Excessively high AMH, in contrast, was at all ages associated with spiking miscarriage rates. CONCLUSIONS: At varying peripheral serum concentrations, AMH, thus, demonstrates hithero unknown and contradictory effects on IVF outcomes, deserving at different concentrations investigation as a potential therapeutic agent, with pregnancy-supporting and pregnancy-interrupting properties.


Asunto(s)
Hormona Antimülleriana/metabolismo , Embrión de Mamíferos/citología , Fertilización In Vitro , Hormona Folículo Estimulante/metabolismo , Adulto , Factores de Edad , Estudios de Cohortes , Parto Obstétrico , Femenino , Humanos , Modelos Biológicos , Embarazo , Pronóstico , Resultado del Tratamiento
8.
Front Pharmacol ; 14: 1285779, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38155905

RESUMEN

Preterm birth is the leading cause of infant morbidity and mortality. There has been an interest in developing prostaglandin F2α (PGF2α) antagonists as a new treatment for preterm birth, although much of the rationale for their use is based on studies in rodents where PGF2α initiates labour by regressing the corpus luteum and reducing systemic progesterone concentrations. How PGF2α antagonism would act in humans who do not have a fall in systemic progesterone remains unclear. One possibility, in addition to an acute stimulation of contractions, is a direct alteration of the myometrial smooth muscle cell state towards a pro-labour phenotype. In this study, we developed an immortalised myometrial cell line, MYLA, derived from myometrial tissue obtained from a pregnant, non-labouring patient, as well as a novel class of PGF2α receptor (FP) antagonist. We verified the functionality of the cell line by stimulation with PGF2α, resulting in Gαq-specific coupling and Ca2+ release, which were inhibited by FP antagonism. Compared to four published FP receptor antagonists, the novel FP antagonist N582707 was the most potent compound [Fmax 7.67 ± 0.63 (IC50 21.26 nM), AUC 7.30 ± 0.32 (IC50 50.43 nM), and frequency of Ca2+ oscillations 7.66 ± 0.41 (IC50 22.15 nM)]. RNA-sequencing of the MYLA cell line at 1, 3, 6, 12, 24, and 48 h post PGF2α treatment revealed a transforming phenotype from a fibroblastic to smooth muscle mRNA profile. PGF2α treatment increased the expression of MYLK, CALD1, and CNN1 as well as the pro-labour genes OXTR, IL6, and IL11, which were inhibited by FP antagonism. Concomitant with the inhibition of a smooth muscle, pro-labour transition, FP antagonism increased the expression of the fibroblast marker genes DCN, FBLN1, and PDGFRA. Our findings suggest that in addition to the well-described acute contractile effect, PGF2α transforms myometrial smooth muscle cells from a myofibroblast to a smooth muscle, pro-labour-like state and that the novel compound N582707 has the potential for prophylactic use in preterm labour management beyond its use as an acute tocolytic drug.

9.
Endocrinology ; 163(5)2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35396990

RESUMEN

In females, reproductive success is dependent on the expression of a number of genes regulated at different levels, one of which is through epigenetic modulation. How a specific epigenetic modification regulates gene expression and their downstream effect on ovarian function are important for understanding the female reproductive process. The trimethylation of histone3 at lysine27 (H3K27me3) is associated with gene repression. JMJD3 (or KDM6b), a jumonji domain-containing histone demethylase specifically catalyzes the demethylation of H3K27me3, that positively influences gene expression. This study reports that the expression of JMJD3 specifically in the ovarian granulosa cells (GCs) is critical for maintaining normal female fertility. Conditional deletion of Jmjd3 in the GCs results in a decreased number of total healthy follicles, disrupted estrous cycle, and increased follicular atresia culminating in subfertility and premature ovarian failure. At the molecular level, the depletion of Jmjd3 and RNA-seq analysis reveal that JMJD3 is essential for mitochondrial function. JMJD3-mediated reduction of H3K27me3 induces the expression of Lif (Leukemia inhibitory factor) and Ctnnb1 (ß-catenin), that in turn regulate the expression of key mitochondrial genes critical for the electron transport chain. Moreover, mitochondrial DNA content is also significantly decreased in Jmjd3 null GCs. Additionally, we have uncovered that the expression of Jmjd3 in GCs decreases with age, both in mice and in humans. Thus, in summary, our studies highlight the critical role of JMJD3 in nuclear-mitochondrial genome coordination that is essential for maintaining normal ovarian function and female fertility and underscore a potential role of JMJD3 in female reproductive aging.


Asunto(s)
Atresia Folicular , Histonas , Histona Demetilasas con Dominio de Jumonji/metabolismo , Animales , Femenino , Fertilidad/genética , Histonas/metabolismo , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Ratones , Ovario/metabolismo
10.
Endocrinology ; 163(10)2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35933634

RESUMEN

In women, excess androgen causes polycystic ovary syndrome (PCOS), a common fertility disorder with comorbid metabolic dysfunctions including diabetes, obesity, and nonalcoholic fatty liver disease. Using a PCOS mouse model, this study shows that chronic high androgen levels cause hepatic steatosis while hepatocyte-specific androgen receptor (AR)-knockout rescues this phenotype. Moreover, through RNA-sequencing and metabolomic studies, we have identified key metabolic genes and pathways affected by hyperandrogenism. Our studies reveal that a large number of metabolic genes are directly regulated by androgens through AR binding to androgen response element sequences on the promoter region of these genes. Interestingly, a number of circadian genes are also differentially regulated by androgens. In vivo and in vitro studies using a circadian reporter [Period2::Luciferase (Per2::LUC)] mouse model demonstrate that androgens can directly disrupt the hepatic timing system, which is a key regulator of liver metabolism. Consequently, studies show that androgens decrease H3K27me3, a gene silencing mark on the promoter of core clock genes, by inhibiting the expression of histone methyltransferase, Ezh2, while inducing the expression of the histone demethylase, JMJD3, which is responsible for adding and removing the H3K27me3 mark, respectively. Finally, we report that under hyperandrogenic conditions, some of the same circadian/metabolic genes that are upregulated in the mouse liver are also elevated in nonhuman primate livers. In summary, these studies not only provide an overall understanding of how hyperandrogenism associated with PCOS affects liver gene expression and metabolism but also offer insight into the underlying mechanisms leading to hepatic steatosis in PCOS.


Asunto(s)
Hiperandrogenismo , Enfermedad del Hígado Graso no Alcohólico , Síndrome del Ovario Poliquístico , Andrógenos/metabolismo , Andrógenos/farmacología , Animales , Modelos Animales de Enfermedad , Epigénesis Genética , Femenino , Histonas/metabolismo , Humanos , Hiperandrogenismo/complicaciones , Ratones , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Enfermedad del Hígado Graso no Alcohólico/genética , Síndrome del Ovario Poliquístico/metabolismo
11.
J Biol Chem ; 285(37): 28787-95, 2010 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-20628053

RESUMEN

Although transcriptional effects of androgens have been extensively studied, mechanisms regulating transcription-independent (nongenomic) androgen actions are poorly understood. Previously, we have shown that paxillin, a multidomain adaptor protein, is a critical regulator of testosterone-induced MAPK-signaling during Xenopus oocyte maturation. Here we examine the nongenomic effects of dihydrotestosterone (DHT) in prostate cancer cells, focusing on how paxillin mediates Erk signaling and downstream physiologic actions. We show that in LnCAP cells DHT functions as a growth factor that indirectly activates the EGF-receptor (EGFR) via androgen receptor binding and matrix metalloproteinase-mediated release of EGFR ligands. Interestingly, siRNA-mediated knockdown of paxillin expression in androgen-dependent LnCAP cells as well as in androgen-independent PC3 cells abrogates DHT- and/or EGF-induced Erk signaling. Furthermore, EGFR-induced Erk activation requires Src-mediated phosphorylation of paxillin on tyrosines 31/118. In contrast, paxillin is not required for PKC-induced Erk signaling. However, Erk-mediated phosphorylation of paxillin on serines 83/126/130 is still needed for both EGFR and PKC-mediated cellular proliferation. Thus, paxillin serves as a specific upstream regulator of Erk in response to receptor-tyrosine kinase signaling but as a general regulator of downstream Erk actions regardless of agonist. Importantly, Erk-mediated serine phosphorylation of paxillin is also required for DHT-induced prostate-specific antigen mRNA expression in LnCAP cells as well as EGF-induced cyclin D1 mRNA expression in PC3 cells, suggesting that paxillin may regulate prostate cancer proliferation by serving as a liaison between extra-nuclear kinase signaling and intra-nuclear transcriptional signals. Thus, paxillin may prove to be a novel diagnostic or therapeutic target in prostate cancer.


Asunto(s)
Andrógenos/farmacología , Proliferación Celular/efectos de los fármacos , Dihidrotestosterona/farmacología , Factor de Crecimiento Epidérmico/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Paxillin/metabolismo , Neoplasias de la Próstata/metabolismo , Línea Celular Tumoral , Receptores ErbB/genética , Receptores ErbB/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Masculino , Paxillin/genética , Fosforilación/efectos de los fármacos , Fosforilación/genética , Antígeno Prostático Específico/biosíntesis , Antígeno Prostático Específico/genética , Neoplasias de la Próstata/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética
12.
J Endocrinol ; 249(3): R53-R64, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33764313

RESUMEN

In recent years, androgens have emerged as critical regulators of female reproduction and women's health in general. While high levels of androgens in women are associated with polycystic ovary syndrome (PCOS), recent evidence suggests that a certain amount of direct androgen action through androgen receptor is also essential for normal ovarian function. Moreover, prenatal androgen exposure has been reported to cause developmental reprogramming of the fetus that manifests into adult pathologies, supporting the Developmental Origins of Health and Disease (DOHaD) hypothesis. Therefore, it has become imperative to understand the underlying mechanism of androgen actions and its downstream effects under normal and pathophysiological conditions. Over the years, there has been a lot of studies on androgen receptor function as a transcriptional regulator in the nucleus as well as androgen-induced rapid extra-nuclear signaling. Conversely, new evidence suggests that androgen actions may also be mediated through epigenetic modulation involving both the nuclear and extra-nuclear androgen signaling. This review focuses on androgen-induced epigenetic modifications in female reproduction, specifically in the ovary, and discusses emerging concepts, latest perceptions, and highlight the areas that need further investigation.


Asunto(s)
Andrógenos/farmacología , Epigénesis Genética , Regulación de la Expresión Génica/efectos de los fármacos , Ovario/efectos de los fármacos , Ovario/metabolismo , Femenino , Humanos , Infertilidad Femenina
13.
Endocrinology ; 162(12)2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34597389

RESUMEN

Altered nutrition or intrauterine exposure to various adverse conditions during fetal development or earlier in a mother's life can lead to epigenetic changes in fetal tissues, predisposing those tissues to diseases that manifest when offspring become adults. An example is a maternal obesity associated with gestational diabetes (GDM), where fetal exposure to a hyperglycemic, hyperinsulinemic, and/or hyperlipidemic gestational environment can provoke epigenetic changes that predispose offspring to various diseased conditions later in life. While it is now well established that offspring exposed to GDM have an increased risk of developing obesity, metabolic disorders, and/or cardiovascular disease in adult life, there are limited studies assessing the reproductive health of these offspring. This mini-review discusses the long-term effect of in utero exposure to GDM-associated adverse prenatal environment on the reproductive health of the offspring. Moreover, using evidence from various animal models and human epidemiological studies, this review offers molecular insight and understanding of how epigenetic reprogramming of genes culminates in reproductive dysfunction and the development of subfertility or infertility later in adult life.


Asunto(s)
Diabetes Gestacional/fisiopatología , Infertilidad/etiología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Animales , Diabetes Gestacional/patología , Femenino , Fertilidad/fisiología , Humanos , Infertilidad/embriología , Madres , Embarazo , Efectos Tardíos de la Exposición Prenatal/etiología , Útero/patología , Útero/fisiología
14.
Sci Rep ; 11(1): 20956, 2021 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-34697337

RESUMEN

The vagina plays a critical role in supporting the pelvic organs and loss of support leads to pelvic organ prolapse. It is unknown what microstructural changes influence prolapse progression nor how decreased elastic fibers contributes to vaginal remodeling and smooth muscle contractility. The objective for this study was to evaluate the effect of fibulin-5 haploinsufficiency, and deficiency with progressive prolapse on the biaxial contractile and biomechanical function of the murine vagina. Vaginas from wildtype (n = 13), haploinsufficient (n = 13), and deficient mice with grade 1 (n = 9) and grade 2 or 3 (n = 9) prolapse were explanted for biaxial contractile and biomechanical testing. Multiaxial histology (n = 3/group) evaluated elastic and collagen fiber microstructure. Western blotting quantified protein expression (n = 6/group). A one-way ANOVA or Kruskal-Wallis test evaluated statistical significance. Pearson's or Spearman's test determined correlations with prolapse grade. Axial contractility decreased with fibulin-5 deficiency and POP (p < 0.001), negatively correlated with prolapse grade (ρ = - 0.80; p < 0.001), and positively correlated with muscularis elastin area fraction (ρ = - 0.78; p = 0.004). Circumferential (ρ = 0.71; p < 0.001) and axial (ρ = 0.69; p < 0.001) vaginal wall stresses positively correlated with prolapse grade. These findings demonstrated that fibulin-5 deficiency and prolapse progression decreased vaginal contractility and increased vaginal wall stress. Future work is needed to better understand the processes that contribute to prolapse progression in order to guide diagnostic, preventative, and treatment strategies.


Asunto(s)
Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Prolapso Uterino/fisiopatología , Vagina/fisiopatología , Animales , Fenómenos Biomecánicos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Haploinsuficiencia , Humanos , Ratones , Estrés Mecánico , Prolapso Uterino/genética , Prolapso Uterino/metabolismo , Vagina/metabolismo
15.
Breast Cancer Res ; 12(3): 109, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20561377

RESUMEN

The diversity of membrane-initiated progesterone actions has made characterization and establishment of its biological importance a complicated endeavor. A new study by Zuo and colleagues shows that progesterone via endogenous membrane progesterone receptor-alpha acts as a negative regulator of proliferation and epithelial to mesenchymal transition in a breast cancer cell line. These progesterone-mediated actions appear to be regulated through epidermal growth factor receptor and phosphatidylinositol 3-kinase signaling localized in caveolae. Moreover, the study shows expression of membrane progesterone receptor-alpha in benign and malignant breast cancer tissues. These data bring forth novel concepts with regard to progesterone actions in the breast; however, further work is warranted to fully characterize the physiologic actions of extra-nuclear progesterone signaling in the breast.


Asunto(s)
Neoplasias de la Mama/metabolismo , Membrana Celular/metabolismo , Mesodermo/metabolismo , Progesterona/fisiología , Receptores de Progesterona/metabolismo , Transducción de Señal/fisiología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Humanos , Mesodermo/patología , Progestinas/fisiología , Receptores de Progesterona/antagonistas & inhibidores
16.
Mol Cell Endocrinol ; 501: 110655, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-31756424

RESUMEN

Precise timing in hormone release from the hypothalamus, the pituitary and ovary is critical for fertility. Hormonal release patterns of the reproductive axis are regulated by a feedback loop within the hypothalamic-pituitary-gonadal (HPG) axis. The timing and rhythmicity of hormone release and tissue sensitivity in the HPG axis is regulated by circadian clocks located in the hypothalamus (suprachiasmatic nucleus, kisspeptin and GnRH neurons), the pituitary (gonadotrophs), the ovary (theca and granulosa cells), the testis (Leydig cells), as well as the uterus (endometrium and myometrium). The circadian clocks integrate environmental and physiological signals to produce cell endogenous rhythms generated by a transcriptional-translational feedback loop of transcription factors that are collectively called the "molecular clock". This review specifically focuses on the contribution of molecular clock transcription factors in regulating hormone release patterns in the reproductive axis, with an emphasis on the female reproductive system. Specifically, we discuss the contributions of circadian rhythms in distinct neuronal populations of the female hypothalamus, the molecular clock in the pituitary and its overall impact on female and male fertility.


Asunto(s)
Proteínas CLOCK/genética , Relojes Circadianos/genética , Ritmo Circadiano/genética , Reproducción/genética , Animales , Relojes Circadianos/fisiología , Ritmo Circadiano/fisiología , Fertilidad/genética , Humanos , Hipotálamo/fisiología , Hipófisis/fisiología , Reproducción/fisiología
17.
Biol Reprod ; 81(3): 580-6, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19439726

RESUMEN

We demonstrated previously a negative association of granulosa cell cocaine- and amphetamine-regulated transcript (CARTPT) expression with follicle health status and inhibitory effects of the mature CARTPT peptide (CART) on follicle-stimulating hormone (FSH) signal transduction in vitro, resulting in reduced bovine granulosa cell CYP19A1 mRNA and estradiol production. The objectives of this study were to investigate temporal regulation of granulosa cell CARTPT expression (granulosa cell mRNA and follicular fluid CART peptide concentrations) during follicular waves, CART regulation of androstenedione production (precursor for estradiol biosynthesis) by thecal tissue collected at specific stages of a follicular wave, FSH regulation of granulosa cell CARTPT mRNA expression, and the ability of CART to inhibit granulosa cell estradiol production and CYP19A1 mRNA expression when administered in vivo. CART concentrations in healthy, estrogen-active follicles (estradiol greater than progesterone in follicular fluid) decreased after dominant follicle selection, and CARTPT mRNA was lower in healthy, estrogen-active versus estrogen-inactive atretic follicles (progesterone greater than estradiol) collected at the predeviation and early dominance stages. CART treatment reduced luteinizing hormone-induced androstenedione production by thecal tissue collected at predeviation and early dominance stages but not at later stages of a follicular wave. The FSH or insulin-like growth factor 1 treatment in vitro reduced granulosa cell CARTPT mRNA in a dose-dependent fashion. Administration of CART in vivo into follicles at the early dominance stage reduced follicular fluid estradiol concentrations and granulosa cell CYP19A1 mRNA. Collectively, results support a potential stage-specific regulatory role for CART in negative regulation of estradiol production associated with selection of the dominant follicle.


Asunto(s)
Bovinos , Estradiol/metabolismo , Células de la Granulosa/metabolismo , Proteínas del Tejido Nervioso/fisiología , Folículo Ovárico/citología , Androstenodiona/metabolismo , Animales , Aromatasa/genética , Aromatasa/metabolismo , Bovinos/genética , Bovinos/metabolismo , Bovinos/fisiología , Células Cultivadas , Femenino , Hormona Folículo Estimulante/farmacología , Líquido Folicular/química , Líquido Folicular/metabolismo , Regulación Enzimológica de la Expresión Génica , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/fisiología , Factor I del Crecimiento Similar a la Insulina/farmacología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Folículo Ovárico/metabolismo , Folículo Ovárico/fisiología
19.
Mol Endocrinol ; 22(12): 2655-76, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18818282

RESUMEN

Pleiotropic actions of cocaine- and amphetamine-regulated transcript (CART) are well described in the central nervous system and periphery, but the intracellular mechanisms mediating biological actions of CART are poorly understood. Although CART is not expressed in mouse ovaries, we have previously established CART as a novel intracellular regulator of estradiol production in bovine granulosa cells. We demonstrated that inhibitory actions of CART on estradiol production are mediated through inhibition of FSH-induced cAMP accumulation, Ca(2+) influx, and aromatase mRNA expression via a G(o/i)-dependent pathway. We also reported that FSH-induced estradiol production is dependent on Erk1/2 and Akt signaling, and CART may regulate other signaling proteins downstream of cAMP essential for estradiol production. Here, we demonstrate that CART is a potent inhibitor of FSH-stimulated Erk1/2 and Akt signaling and the mechanisms involved. Transient CART stimulation of bovine granulosa cells shortens the duration of FSH-induced Erk1/2 and Akt signaling whereas a prolonged (24 h) CART treatment blocks Erk1/2 and Akt activation in response to FSH. This CART-induced accelerated termination of Erk1/2 and Akt signaling is mediated both by induced expression and impaired ubiquitin-mediated proteasome degradation of dual specific phosphatase 5 (DUSP5) and protein phosphatase 2A. Results also support existence of a negative feedback loop in which CART via a G(o/i)-MAPK kinase dependent pathway activates Erk1/2, and the latter induces DUSP5 expression. Moreover, small interfering RNA mediated ablation of DUSP5 and/or protein phosphatase 2A prevents the CART-induced early termination of Erk1/2 and Akt signaling. Results provide novel insight into the intracellular mechanism of action of CART in regulation of FSH-induced MAPK signaling.


Asunto(s)
Hormona Folículo Estimulante/farmacología , Células de la Granulosa/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/metabolismo , Proteínas del Tejido Nervioso/fisiología , Proteína Oncogénica v-akt/metabolismo , Animales , Bovinos , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/enzimología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/antagonistas & inhibidores , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/genética , Modelos Biológicos , Proteínas del Tejido Nervioso/farmacología , Proteína Quinasa C/metabolismo , Interferencia de ARN/efectos de los fármacos , Interferencia de ARN/fisiología , ARN Interferente Pequeño/farmacología , Transducción de Señal/efectos de los fármacos
20.
Endocrinology ; 160(7): 1684-1700, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31150057

RESUMEN

Intrauterine exposure to various adverse conditions during fetal development can lead to epigenetic changes in fetal tissues, predisposing those tissues to disease conditions later in life. An example is gestational diabetes (GD), where the offspring has a higher risk of developing obesity, metabolic disorders, or cardiovascular disease in adult life. In this study, using two well-established GD (streptozotocin- and high-fat and high-sugar-induced) mouse models, we report that female offspring from GD dams are predisposed toward fertility problems later in life. This predisposition to fertility problems is due to altered ovarian expression of a peptide called cocaine- and amphetamine-regulated transcript (CART), which is known to negatively affect folliculogenesis and is induced by elevated leptin levels. Results show that the underlying cause of this altered expression is due to fetal epigenetic modifications involving glucose- and insulin-induced miRNA, miR-101, and the phosphatidylinositol 3-kinase/Akt pathway. These signaling events regulate Ezh2, a histone methyltransferase that promotes H3K27me3, a gene-repressive mark, and CBP/p300, a histone acetyltransferase that promotes H3K27ac, a transcription activation mark, in the fetal ovary. Moreover, the CART promoter has depleted 5-methylcytosine (5mC) and enriched 5-hydroxymethylcytosine (5hmC) levels. The depletion of H3K27me3 and 5mC repressive marks and subsequent increase in H3K27ac and 5hmC gene-activating marks convert the Cartpt promoter to a "superpromoter." This makes the Cartpt promoter more sensitive to leptin levels that predispose the GD offspring to fertility problems. Therefore, this study provides a mechanistic insight about fetal epigenome reprogramming that manifests to ovarian dysfunction and subfertility later in adult life.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Gestacional/metabolismo , Epigénesis Genética , Infertilidad/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Ovario/metabolismo , Regiones Promotoras Genéticas , Animales , Metilación de ADN , Diabetes Mellitus Experimental/genética , Diabetes Gestacional/genética , Femenino , Histonas/metabolismo , Infertilidad/genética , Ratones , Proteínas del Tejido Nervioso/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Embarazo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA