Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Mol Cell Endocrinol ; 587: 112214, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38537882

RESUMEN

Both male and female reproductive functions are impacted by altered gonadotrophin secretion and action, which may also influence the development of endocrine tumours. To ascertain if chronic hypersecretion of human chorionic gonadotropin (hCG) contributes to the development of gonadal tumours, double transgenic (TG) mice that overexpress hCGα- and ß-subunits were analysed. By the age of two months, ovarian tumours with characteristics of teratomas developed with 100% penetrance. Teratomas were also seen in wild-type ovaries orthotopically transplanted into TG mice, demonstrating an endocrine/paracrine mechanism for the hCG-induced ovarian tumorigenesis. Both in vitro and in vivo experiments showed oocyte parthenogenetic activation in TG females. In addition, ovaries showed reduced ovulatory gene expression, inhibited ERK1/2 phosphorylation, and impaired cumulus cell expansion. Hence, persistently high endocrine hCG activity causes parthenogenetic activation and development of ovarian teratomas, along with altered follicle development and impaired ERK1/2 signalling, offering a novel mechanism associated with the molecular pathogenesis of ovarian teratomas.


Asunto(s)
Neoplasias Ováricas , Teratoma , Ratones , Animales , Masculino , Femenino , Humanos , Lactante , Ratones Transgénicos , Gonadotropina Coriónica/farmacología , Oocitos , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología
2.
Gen Comp Endocrinol ; 336: 114247, 2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-36858273

RESUMEN

The hypothalamic-pituitary-gonadal axis plays a fundamental role in the endocrine regulation of the reproductive function in mammals. Any change in the function of the participating hormones or their receptors can lead to alterations in sexual differentiation, the onset of puberty, infertility, cancer development, and other dysfunctions. In this study, we analyzed the influence of persistently elevated levels of the human chorionic gonadotropin hormone (hCG), a powerful agonist of pituitary luteinizing hormone (LH), on the reproductive axis of female mice. As a consequence of chronic hCG hypersecretion through a global expression of the hCGbeta-subunit in transgenic (TG) female mice, a series of events perturbed the prepubertal to juvenile transition. The imbalance in gonadotropin action was first manifested by precocious puberty and alterations in gonadal hormone production, with the consequent ovarian function disruption and infertility in adulthood. The expansion of cumulus cells in vivo and in vitro, ovulatory capacity, and gene expression of ovulation-related marker genes after hormone stimulation were normal in 3-week-old TG females. However, the expression of genes related to steroidogenesis and luteinization such as Lhcgr, Prlr, and the steroidogenic enzymes Cyp11a1, Cyp17a1, and Cyp19a1 were significantly elevated in the TG females. This study demonstrates that the excessive secretion of hCG in concert with high prolactin, induced premature luteinization, and enhanced ovarian steroidogenesis, as was shown by the up-regulation of luteal cell markers and progesterone synthesis in the TG mice. Furthermore, progressively impaired reproductive function of the TG females occurred from the peripubertal stage to adulthood, thus culminating in infertility.


Asunto(s)
Gonadotropina Coriónica , Infertilidad , Humanos , Ratones , Femenino , Animales , Gonadotropina Coriónica/farmacología , Gonadotropina Coriónica Humana de Subunidad beta/genética , Gonadotropina Coriónica Humana de Subunidad beta/metabolismo , Ratones Transgénicos , Luteinización , Mamíferos/metabolismo
4.
Endocr Relat Cancer ; 29(6): 359-373, 2022 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-35324456

RESUMEN

Among pituitary adenomas, prolactinomas are the most frequently diagnosed (about 50%). Dopamine agonists are generally effective in the treatment of prolactinomas. However, a subset of about 25% of patients does not respond to these agents. The management of drug-resistant prolactinomas remains a challenge for endocrinologists and new inhibitory treatments are needed. Pituitary activins inhibit lactotroph function. Its expression and action were found reduced in animal models of lactotroph hyperplasia (female mice overexpressing the B subunit of the human chorionic gonadotrophin and female mice knockout for dopamine receptor type 2). In these models, an oophorectomy avoids prolactinoma development. Hormonal replacement with oestradiol and/or progesterone is not enough to reach the tumor size observed in transgenic females. We postulated that the loss of gonadal inhibins after an oophorectomy contributes to prevent hyperplasia development. Here, we demonstrated that an oophorectomy at 2 months age recovers the following in adulthood: (i) pituitary activin expression, (ii) activin receptor expression specifically in lactotroph population, (iii) activin biological activity in lactotrophs with a concomitant reduction of Pit-1 expression. To summarize, when an oophorectomy is performed, inhibins are lost and the inhibitory action of pituitary activins on lactotroph population is recovered, helping to prevent lactotroph hyperplasia development. These results emphasize the importance of the inhibitory action of activins on lactotroph function, positioning activins as a good therapeutic target for the treatment of resistant prolactinomas.


Asunto(s)
Lactotrofos , Neoplasias Hipofisarias , Prolactinoma , Activinas/metabolismo , Adulto , Animales , Femenino , Humanos , Hiperplasia , Inhibinas/metabolismo , Inhibinas/uso terapéutico , Lactotrofos/metabolismo , Lactotrofos/patología , Ratones , Ovariectomía , Neoplasias Hipofisarias/metabolismo , Prolactina/metabolismo , Prolactinoma/metabolismo , Prolactinoma/prevención & control
5.
Reproduction ; 161(5): 539-548, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33730689

RESUMEN

Heterospecific embryo transfer of an endangered species has been carried out using recipients from related domestic females. Aggregation of an embryo from an endangered species with a tetraploid embryo from the species to be transferred could improve the development of pregnancy to term. The main objective of the present study was to analyze embryo aggregation in domestic cat model using hybrid embryos. For this purpose, we compared in vitro development of synchronic (Sync) or asynchronic (Async) and asynchronic with a tetraploid (Async4n) aggregation of domestic cat IVF embryos. Furthermore, aggregated blastocyst quality was analyzed by evaluation of the total cell number, cell allocation by mitotrackers staining of embryonic cells, expression of Oct4, Nanog, Sox2, Cdx2 genes, number of OCT4+ nuclei, and presence of DNA fragmentation. Additionally, the developmental rates of Async4n aggregation of domestic cat with Leopardus geoffroyi hybrid (hLg) embryos were evaluated. Async aggregation increased blastocyst cell number and the number of OCT4+ nuclei as compared to non-aggregated diploid (2n) and tetraploid (4n) embryos. Moreover, blastocysts produced by Async4n aggregation showed reduced rates of fragmented DNA. No differences were found in the expression of the pluripotent genes, with exception of the Cdx2 expression, which was higher in 4n and aggregated embryos as compared to the control group. Interestingly, hybrids embryos derived by Async4n aggregation with domestic cat embryos had similar rates of blastocysts development as the control. Altogether, the findings support the use of two-cell-fused embryos to generate tetraploid blastomeres and demonstrate that Async4n aggregation generates good quality embryos.


Asunto(s)
Blastómeros/fisiología , Fusión Celular , Embrión de Mamíferos/citología , Desarrollo Embrionario , Fertilización In Vitro/veterinaria , Tetraploidía , Animales , Blastómeros/citología , Gatos , Transferencia de Embrión , Embrión de Mamíferos/metabolismo , Femenino , Masculino , Panthera , Embarazo
6.
Endocr Relat Cancer ; 26(5): 497-510, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30856609

RESUMEN

Membrane progesterone receptors are known to mediate rapid nongenomic progesterone effects in different cell types. Recent evidence revealed that mPRα is highly expressed in the rat pituitary, being primarily localized in lactotrophs, acting as an intermediary of P4-inhibitory actions on prolactin secretion. The role of mPRs in prolactinoma development remains unclear. We hypothesize that mPR agonists represent a novel tool for hyperprolactinemia treatment. To this end, pituitary expression of mPRs was studied in three animal models of prolactinoma. Expression of mPRs and nuclear receptor was significantly decreased in tumoral pituitaries compared to normal ones. However, the relative proportion of mPRα and mPRß was highly increased in prolactinomas. Interestingly, the selective mPR agonist (Org OD 02-0) significantly inhibited PRL release in both normal and tumoral pituitary explants, displaying a more pronounced effect in tumoral tissues. As P4 also regulates PRL secretion indirectly, by acting on dopaminergic neurons, we studied mPR involvement in this effect. We found that the hypothalamus has a high expression of mPRs. Interestingly, both P4 and OrgOD 02-0 increased dopamine release in hypothalamus explants. Moreover, in an in vivo treatment, that allows both, pituitary and hypothalamus actions, the mPR agonist strongly reduced the hyperprolactinemia in transgenic females carrying prolactinoma. Finally, we also found and interesting gender difference: males express higher levels of pituitary mPRα/ß, a sex that does not develop prolactinoma in these mice models. Taken together, these findings suggest mPRs activation could represent a novel tool for hyperprolactinemic patients, especially those that present resistance to dopaminergic drugs.


Asunto(s)
Neoplasias Hipofisarias/prevención & control , Progesterona/farmacología , Prolactina/metabolismo , Prolactinoma/prevención & control , Receptores de Dopamina D2/fisiología , Receptores de Progesterona/agonistas , Animales , Gonadotropina Coriónica Humana de Subunidad beta/genética , Gonadotropina Coriónica Humana de Subunidad beta/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Neoplasias Hipofisarias/etiología , Neoplasias Hipofisarias/patología , Prolactinoma/etiología , Prolactinoma/patología , Ratas , Transducción de Señal
7.
Artículo en Inglés | MEDLINE | ID: mdl-30761084

RESUMEN

The Follicle-Stimulating Hormone plays an important role in the regulation of gametogenesis. It is synthesized and secreted as a family of glycoforms with differing oligosaccharide structure, biological action, and half-life. The presence of these oligosaccharides is absolutely necessary for the full expression of hormone bioactivity at the level of the target cell. The endocrine milieu modulates the glycosylation of this hormone. During male sexual development a progressive increase in FSH sialylation and in the proportion of glycoforms bearing complex oligosaccharides are the main features in this physiological condition. In late puberty, FSH oligosaccharides are largely processed in the medial- and trans-Golgi cisternae of the gonadotrope and remain without changes throughout adult life. In experimental models, the absence of gonads severely affects FSH sialylation; androgen administration is able to restore the characteristics observed under physiological conditions. The expression of ST6 beta-galactoside alpha-2,6-sialyltransferase 1 is hormonally regulated in the male rat; it decreases after short periods of castration but increases markedly at longer periods of androgen deprivation. Although ST3 beta-galactoside alpha-2,3-sialyltransferase 3 is expressed in the male rat pituitary it is not influenced by changes in the endocrine milieu. The oligosaccharide structure of FSH has an impact on the Sertoli cell endocrine activity. In more advanced stages of Sertoli cell maturation, both sialylation and complexity of the oligosaccharides are involved in the regulation of inhibin B production; moreover, FSH glycoforms bearing incomplete oligosaccharides may enhance the stimulatory effect exerted by gonadal growth factors. In this review, we discuss available information on variation of FSH glycosylation and its hormonal regulation under different physiological and experimental conditions, as well as the effect on Sertoli cell endocrine activity.

8.
Front Neuroendocrinol ; 50: 118-122, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29074127

RESUMEN

Prolactinomas are the most frequent functioning pituitary adenomas, and sex differences in tumor size, behavior and incidence have been described. These differences have been associated with earlier diagnosis in woman, as well as with serum estradiol levels. Experimental models of prolactinomas in rodents also show a higher incidence in females, and recent findings suggest that gender differences in the transforming growth factor beta 1 (TGFß1) system might be involved in the sex-specific development of prolactinomas in these models. The aim of this review is to summarize the literature supporting the important role of TGFß1 as a local modulator of pituitary lactotroph function and to provide recent evidence for TGFß1 involvement in the sex differences found in prolactinoma development in animal models.


Asunto(s)
Neoplasias Hipofisarias/metabolismo , Prolactinoma/metabolismo , Caracteres Sexuales , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Femenino , Humanos , Masculino
9.
J Endocrinol ; 232(3): 535-546, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28096433

RESUMEN

Female transgenic mice that overexpress the human chorionic gonadotrophin ß subunit (hCGß+) develop prolactinomas, whereas hCGß+ males do not. The high levels of circulating hCG induce massive luteinization in the ovary of hCGß+ females, and progesterone becomes the primary steroid hormone produced, but estradiol remains at physiological level. The involvement of high levels of progesterone in lactotroph proliferation is not clearly understood; hence, the pathogenesis of prolactinomas in hCGß+ females remains unclear. TGFß1 is an inhibitor of lactotroph function, and the reduced TGFß1 activity found in prolactinomas has been proposed to be involved in tumor development. The aim of the present work was to study the role of TGFß1 in the gender-specific development of prolactinomas in hCGß+ mice. We compared the expression of different components of the pituitary TGFß1 system in males and females in this model. We found reduced TGFß1 levels, reduced expression of TGFß1 target genes, TGFß1 receptors, Ltbp1, Smad4 and Smad7 in hCGß+ female pituitaries. However, no differences were found between the transgenic and wild-type male pituitaries. We postulate that decreased pituitary TGFß1 activity in hCGß+ females is involved in the development of prolactinomas. In fact, we demonstrated that an in vivo treatment carried out for increasing pituitary TGFß1 activity, was successful in reducing the prolactinoma development, and the hyperprolactinemia in hCGß+ females. Moreover, the stronger TGFß1 system found in males could protect them from excessive lactotroph proliferation. Sex differences in the regulation of the pituitary TGFß1 system could explain gender differences in the incidence of prolactinoma.


Asunto(s)
Gonadotropina Coriónica Humana de Subunidad beta/metabolismo , Hipófisis/metabolismo , Neoplasias Hipofisarias/metabolismo , Prolactinoma/metabolismo , Caracteres Sexuales , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Gonadotropina Coriónica Humana de Subunidad beta/genética , Femenino , Proteínas de Unión a TGF-beta Latente/genética , Proteínas de Unión a TGF-beta Latente/metabolismo , Masculino , Ratones , Ratones Transgénicos , Hipófisis/patología , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/patología , Prolactinoma/genética , Prolactinoma/patología , Proteína Smad4/genética , Proteína Smad4/metabolismo , Proteína smad7/genética , Proteína smad7/metabolismo
10.
J Endocrinol ; 230(1): 157-69, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27154336

RESUMEN

The metabolic syndrome is a growing epidemic; it increases the risk for diabetes, cardiovascular disease, fatty liver, and several cancers. Several reports have indicated a link between hormonal imbalances and insulin resistance or obesity. Transgenic (TG) female mice overexpressing the human chorionic gonadotropin ß-subunit (hCGß+ mice) exhibit constitutively elevated levels of hCG, increased production of testosterone, progesterone and prolactin, and obesity. The objective of this study was to investigate the influence of hCG hypersecretion on possible alterations in the glucose and lipid metabolism of adult TG females. We evaluated fasting serum insulin, glucose, and triglyceride levels in adult hCGß+ females and conducted intraperitoneal glucose and insulin tolerance tests at different ages. TG female mice showed hyperinsulinemia, hypertriglyceridemia, and dyslipidemia, as well as glucose intolerance and insulin resistance at 6 months of age. A 1-week treatment with the dopamine agonist cabergoline applied on 5-week-old hCGß+ mice, which corrected hyperprolactinemia, hyperandrogenism, and hyperprogesteronemia, effectively prevented the metabolic alterations. These data indicate a key role of the hyperprolactinemia-induced gonadal dysfunction in the metabolic disturbances of hCGß+ female mice. The findings prompt further studies on the involvement of gonadotropins and prolactin on metabolic disorders and might pave the way for the development of new therapeutic strategies.


Asunto(s)
Gonadotropina Coriónica Humana de Subunidad beta/metabolismo , Intolerancia a la Glucosa/metabolismo , Hiperinsulinismo/metabolismo , Hiperprolactinemia/metabolismo , Hipertrigliceridemia/metabolismo , Resistencia a la Insulina/fisiología , Animales , Glucemia/metabolismo , Cabergolina , Gonadotropina Coriónica Humana de Subunidad beta/genética , Ergolinas/uso terapéutico , Femenino , Intolerancia a la Glucosa/tratamiento farmacológico , Intolerancia a la Glucosa/genética , Hiperinsulinismo/tratamiento farmacológico , Hiperinsulinismo/genética , Hiperprolactinemia/tratamiento farmacológico , Hiperprolactinemia/genética , Hipertrigliceridemia/tratamiento farmacológico , Hipertrigliceridemia/genética , Insulina/sangre , Ratones , Ratones Transgénicos , Prolactina/sangre , Triglicéridos/sangre
11.
Expert Opin Biol Ther ; 15(8): 1183-90, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26160491

RESUMEN

INTRODUCTION: There is continuing need for contraceptives. According to World Health Organization, 210 million pregnancies occur each year, out of which some 80 million are unintended. A vaccine offering privacy and periodic intake would be an attractive proposition. AREAS COVERED: The article is a brief review of three vaccines developed against human chorionic gonadotropin (hCG) with progressively better attributes. Clinical trials have proven in more than one country the complete safety and reversibility of the anti-hCG vaccine(s) in women. Vaccination does not entail any disturbance in levels of reproductive tract hormones of the woman or any disturbance in menstrual regularity and bleeding profiles. Phase II clinical trials show the effective prevention of pregnancy in sexually active women of proven fertility. A recombinant vaccine amenable to industrial production has been developed; it induces substantially higher antibody titers in mice of four different genetic strains than those required to prevent pregnancy in women. Rigorous toxicology studies have been completed on this vaccine in rodents and marmosets. EXPERT OPINION: This unique vaccine, requiring periodic intake and demonstrating no impairment of ovulation, hormonal profiles and menstrual regularity, is on the verge of final clinical trials under the aegis of the Indian Council of Medical Research and should be a valuable addition to the available contraceptives.


Asunto(s)
Gonadotropina Coriónica/antagonistas & inhibidores , Descubrimiento de Drogas/tendencias , Vacunas Anticonceptivas/administración & dosificación , Animales , Antineoplásicos Hormonales/administración & dosificación , Gonadotropina Coriónica/química , Gonadotropina Coriónica/inmunología , Femenino , Humanos , Masculino , Embarazo , Estructura Secundaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/inmunología , Vacunación/métodos , Vacunas Anticonceptivas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología
12.
Reproduction ; 148(4): R63-70, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25030890

RESUMEN

The advent of technologies to genetically manipulate the mouse genome has revolutionised research approaches, providing a unique platform to study the causality of reproductive disorders in vivo. With the relative ease of generating genetically modified (GM) mouse models, the last two decades have yielded multiple loss-of-function and gain-of-function mutation mouse models to explore the role of gonadotrophins and their receptors in reproductive pathologies. This work has provided key insights into the molecular mechanisms underlying reproductive disorders with altered gonadotrophin action, revealing the fundamental roles of these pituitary hormones and their receptors in the hypothalamic-pituitary-gonadal axis. This review will describe GM mouse models of gonadotrophins and their receptors with enhanced or diminished actions, specifically focusing on the male. We will discuss the mechanistic insights gained from these models into male reproductive disorders, and the relationship and understanding provided into male human reproductive disorders originating from altered gonadotrophin action.


Asunto(s)
Gonadotropinas/metabolismo , Infertilidad Masculina/metabolismo , Reproducción , Animales , Hormona Folículo Estimulante/genética , Hormona Folículo Estimulante/metabolismo , Genotipo , Gonadotropinas/genética , Infertilidad Masculina/genética , Infertilidad Masculina/fisiopatología , Hormona Luteinizante/genética , Hormona Luteinizante/metabolismo , Masculino , Ratones , Ratones Transgénicos , Fenotipo , Receptores de HFE/genética , Receptores de HFE/metabolismo , Receptores de HL/genética , Receptores de HL/metabolismo , Reproducción/genética , Transducción de Señal
13.
Mol Cell Endocrinol ; 390(1-2): 102-11, 2014 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-24755422

RESUMEN

Transgenic female mice overexpressing the α- and ß- subunits of human chorionic gonadotropin (hCGαß+) exhibited precocious puberty, as evidenced by early vaginal opening. Chronically elevated hCG in 21-day-old hCGαß+ females stimulated gonadal androgen production, which exerted negative feedback over the endogenous gonadotropin synthesis, and activated the hypothalamic GnRH pulsatility and gene expression. Transgenic females also exhibited elevated hypothalamic aromatization in the preoptic area (POA), which is the sexually-differentiated area that controls the LH surge in adulthood. Ovariectomy at 14 days of age was unable to rescue this phenotype. However, the blockade of androgen action by flutamide from postnatal day 6 onwards reduced the aromatase levels in the POA of hCGαß+ females. Our results suggest that early exposure of females to androgen action during a critical period between postnatal days 6-14 induces sex-specific organizational changes of the brain, which affect the aromatase expression in the POA at the onset of precocious puberty.


Asunto(s)
Gonadotropina Coriónica/metabolismo , Hipotálamo/metabolismo , Pubertad Precoz/metabolismo , Antagonistas de Andrógenos/farmacología , Antagonistas de Andrógenos/uso terapéutico , Animales , Aromatasa/metabolismo , Células Cultivadas , Gonadotropina Coriónica/fisiología , Estradiol/sangre , Femenino , Flutamida/farmacología , Flutamida/uso terapéutico , Hormona Folículo Estimulante/sangre , Expresión Génica , Hormona Liberadora de Gonadotropina/fisiología , Humanos , Ratones Transgénicos , Hipófisis/metabolismo , Pubertad Precoz/tratamiento farmacológico , Testosterona/sangre , Vagina/fisiopatología
14.
Reprod Biol ; 14(1): 9-15, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24607250

RESUMEN

The development of genetically modified animals has been useful to understand the mechanisms involved in the regulation of the gonadotropin function. It is well known that alterations in the secretion of a single hormone is capable of producing profound reproductive abnormalities. Human chorionic gonadotropin (hCG) is a glycoprotein hormone normally secreted by the human placenta, and structurally and functionally it is related to pituitary LH. LH and hCG bind to the same LH/hCG receptor, and hCG is often used as an analog of LH to boost gonadotropin action. There are many physiological and pathological conditions where LH/hCG levels and actions are elevated. In order to understand how elevated LH/hCG levels may impact on the hypothalamic-pituitary-gonadal axis we have developed a transgenic mouse model with chronic hCG hypersecretion. Female mice develop many gonadal and extragonadal phenotypes including obesity, infertility, hyperprolactinemia, and pituitary and mammary gland tumors. This article summarizes recent findings on the mechanisms involved in pituitary gland tumorigenesis and hyperprolactinemia in the female mice hypersecreting hCG, in particular the relationship of progesterone with the hyperprolactinemic condition of the model. In addition, we describe the role of hyperprolactinemia as the main cause of infertility and the phenotypic abnormalities in these mice, and the use of dopamine agonists bromocriptine and cabergoline to normalize these conditions.


Asunto(s)
Gonadotropina Coriónica/metabolismo , Hiperprolactinemia/metabolismo , Receptores de HL/metabolismo , Animales , Gonadotropina Coriónica/genética , Modelos Animales de Enfermedad , Femenino , Hiperprolactinemia/genética , Ratones , Ratones Transgénicos , Hipófisis/metabolismo , Receptores de HL/genética
15.
Neuroendocrinology ; 98(3): 212-23, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24080944

RESUMEN

BACKGROUND/AIMS: Adult mice lacking functional GABAB receptors (GABAB1KO) show altered Gnrh1 and Gad1 expressions in the preoptic area-anterior hypothalamus (POA-AH) and females display disruption of cyclicity and fertility. Here we addressed whether sexual differentiation of the brain and the proper wiring of the GnRH and kisspeptin systems were already disturbed in postnatal day 4 (PND4) GABAB1KO mice. METHODS: PND4 wild-type (WT) and GABAB1KO mice of both sexes were sacrificed; tissues were collected to determine mRNA expression (qPCR), amino acids (HPLC), and hormones (RIA and/or IHC). RESULTS: GnRH neuron number (IHC) did not differ among groups in olfactory bulbs or OVLT-POA. Gnrh1 mRNA (qPCR) in POA-AH was similar among groups. Gnrh1 mRNA in medial basal hypothalamus (MBH) was similar in WTs but was increased in GABAB1KO females compared to GABAB1KO males. Hypothalamic GnRH (RIA) was sexually different in WTs (males > females), but this sex difference was lost in GABAB1KOs; the same pattern was observed when analyzing only the MBH, but not in the POA-AH. Arcuate nucleus Kiss1 mRNA (micropunch-qPCR) was higher in WT females than in WT males and GABAB1KO females. Gad1 mRNA in MBH was increased in GABAB1KO females compared to GABAB1KO males. Serum LH and gonadal estradiol content were also increased in GABAB1KOs. CONCLUSION: We demonstrate that GABABRs participate in the sexual differentiation of the ARC/MBH, because sex differences in several reproductive genes, such as Gad1, Kiss1 and Gnrh1, are critically disturbed in GABAB1KO mice at PND4, probably altering the organization and development of neural circuits governing the reproductive axis.


Asunto(s)
Glutamato Descarboxilasa/deficiencia , Hormona Liberadora de Gonadotropina/deficiencia , Hipotálamo Medio/metabolismo , Kisspeptinas/deficiencia , Precursores de Proteínas/deficiencia , Receptores de GABA-B/deficiencia , Diferenciación Sexual/genética , Animales , Animales Recién Nacidos , Núcleo Arqueado del Hipotálamo/crecimiento & desarrollo , Núcleo Arqueado del Hipotálamo/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Glutamato Descarboxilasa/genética , Hormona Liberadora de Gonadotropina/genética , Hipotálamo Medio/crecimiento & desarrollo , Kisspeptinas/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Precursores de Proteínas/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores de GABA-B/genética
16.
Growth Horm IGF Res ; 23(1-2): 19-28, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23245546

RESUMEN

Postnatal growth exhibits two instances of rapid growth in mice: the first is perinatal and independent of growth hormone (GH), the second is peripuberal and GH-dependent. Signal transducer and activator of transcription 5b (STAT5b) is the main GH-signaling mediator and it is related to IGF1 synthesis and somatic growth. The aim of this work was to assess differential STAT5 sensitivity to GH during the growth period in mouse liver of both sexes. Three representative ages were selected: 1-week-old animals, in the GH-independent phase of growth; 2.5-week-old mice, at the onset of the GH-dependent phase of growth; and 9-week-old young adults. GH-signaling mediators were assessed by immunoblotting, quantitative RT-PCR and immunohistochemistry. GH-induced STAT5 phosphorylation is low at one-week and maximal at 2.5-weeks of age when compared to young adults, accompanied by higher protein content at the onset of growth. Suppressor CIS and phosphatase PTP1B exhibit high levels in one-week animals, which gradually decline, while SOCS2 and SOCS3 display higher levels at adulthood. Nuclear phosphorylated STAT5 is low in one-week animals while in 2.5-week animals it is similar to 9-week control; expression of SOCS3, an early response GH-target gene, mimics this pattern. STAT5 coactivators glucocorticoid receptor (GR) and hepatic nuclear factor 1 (HNF1) abundance is higher in adulthood. Therefore, GH-induced STAT5 signaling presents age-dependent activity in liver, with its maximum coinciding with the onset of GH-dependent phase of growth, accompanied by an age-dependent variation of modulating factors. This work contributes to elucidate the molecular mechanisms implicated in GH responsiveness during growth.


Asunto(s)
Hormona del Crecimiento/metabolismo , Crecimiento y Desarrollo , Hígado/crecimiento & desarrollo , Hígado/metabolismo , Factor de Transcripción STAT5/metabolismo , Factores de Edad , Animales , Núcleo Celular/efectos de los fármacos , Núcleo Celular/genética , Núcleo Celular/metabolismo , Femenino , Hormona del Crecimiento/farmacología , Crecimiento y Desarrollo/efectos de los fármacos , Crecimiento y Desarrollo/genética , Crecimiento y Desarrollo/fisiología , Janus Quinasa 2/metabolismo , Hígado/efectos de los fármacos , Masculino , Ratones , Fosforilación/efectos de los fármacos , Receptores de Somatotropina/genética , Receptores de Somatotropina/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
17.
Endocrinology ; 153(12): 5980-92, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23117930

RESUMEN

Female infertility is often associated with deregulation of hormonal networks, and hyperprolactinemia is one of the most common endocrine disorders of the hypothalamic-pituitary axis affecting the reproductive functions. We have shown previously that transgenic female mice overexpressing human chorionic gonadotropin ß-subunit (hCGß+ mice), and producing elevated levels of bioactive LH/hCG, exhibit increased production of testosterone and progesterone, are overweight and infertile, and develop hyperprolactinemia associated with pituitary lactotrope adenomas in adult age. In the present study, we analyzed the influence of the hyperprolactinemia of hCGß+ females on their reproductive phenotype by treating them with the dopamine agonists, bromocriptine and cabergoline. Long-term bromocriptine treatment of adult mice was effective in the control of obesity, pituitary growth, and disturbances in the hormone profile, demonstrating that hyperprolactinemia was the main cause of the hCGß+ female phenotype. Interestingly, short-term treatment (1 wk) with cabergoline applied on 5-wk-old mice corrected hyperprolactinemia, hyperandrogenism, and hyperprogesteronemia, prevented pituitary overgrowth, normalized gonadal function, and recovered fertility of adult hCGß+ females after hormone-induced and natural ovulation. The same cabergoline treatment in the short term applied on 3-month-old hCGß+ females failed to recover their reproductive function. Hence, we demonstrated that the short-term cabergoline treatment applied at a critical early stage of the phenotype progression effectively prevented the hyperprolactinemia-associated reproductive dysfunction of hCG-overproducing females.


Asunto(s)
Gonadotropina Coriónica/metabolismo , Hiperprolactinemia/complicaciones , Infertilidad/complicaciones , Infertilidad/metabolismo , Animales , Bromocriptina/farmacología , Cabergolina , Modelos Animales de Enfermedad , Ergolinas/farmacología , Femenino , Fertilidad , Regulación de la Expresión Génica , Humanos , Hiperprolactinemia/metabolismo , Ratones , Ratones Transgénicos , Ovulación , Fenotipo , Factores de Tiempo
18.
Rev Endocr Metab Disord ; 12(4): 245-58, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21484328

RESUMEN

During the last two decades a large number of genetically modified mouse lines with altered gonadotropin action have been generated. These mouse lines fall into three categories: the lack-of-function mice, gain-of-function mice, and the mice generated by breeding the abovementioned lines with other disease model lines. The mouse strains lacking gonadotropin action have elucidated the necessity of the pituitary hormones in pubertal development and function of gonads, and revealed the processes from the original genetic defect to the pathological phenotype such as hypo- or hypergonadotropic hypogonadism. Conversely, the strains of the second group depict consequences of chronic gonadotropin action. The lines vary from those expressing constitutively active receptors and those secreting follicle-stimulating hormone (FSH) with slowly increasing amounts to those producing human choriogonadotropin (hCG), amount of which corresponds to 2000-fold luteinizing hormone (LH)/hCG biological activity. Accordingly, the phenotypes diverge from mild anomalies and enhanced fertility to disrupted gametogenesis, but eventually chronic, enhanced and non-pulsatile action of both FSH and LH leads to female and male infertility and/or hyper- and neoplasias in most of the gonadotropin gain-of-function mice. Elevated gonadotropin levels also alter the function of several extra-gonadal tissues either directly or indirectly via increased sex steroid production. These effects include promotion of tumorigenesis in tissues such as the pituitary, mammary and adrenal glands. Finally, the crossbreedings of the current mouse strains with other disease models are likely to uncover the contribution of gonadotropins in novel biological systems, as exemplified by the recent crossbreed of LHCG receptor deficient mice with Alzheimer disease mice.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedades del Sistema Endocrino/genética , Gonadotropinas/genética , Gonadotropinas/fisiología , Ratones Transgénicos , Animales , Enfermedades del Sistema Endocrino/metabolismo , Enfermedades del Sistema Endocrino/patología , Enfermedades del Sistema Endocrino/fisiopatología , Femenino , Gonadotropinas/deficiencia , Gonadotropinas/metabolismo , Humanos , Masculino , Ratones , Ovario/metabolismo , Ovario/patología , Ovario/fisiología , Hipófisis/metabolismo , Hipófisis/patología , Hipófisis/fisiología , Testículo/metabolismo , Testículo/patología , Testículo/fisiología
19.
Mol Cell Endocrinol ; 332(1-2): 78-87, 2011 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-20933053

RESUMEN

Transgenic male mice that express human chorionic gonadotropin (hCG) α and ß subunits constitutively hypersecrete hCG and produce elevated levels of androgens. The aim of this study was to characterize the hypothalamic-pituitary function of these transgenic (hCGαß+) males by focusing on FSH regulation. Serum FSH levels and pituitary mRNA expression of Fshb, Lhb, Cga, Gnrhr and Esr1 were reduced, whereas Fst expression was increased in prepubertal hCGαß+ males as compared with wild-type. In the hypothalamus, Cyp19a1 expression, GnRH concentration and ex-vivo GnRH pulsatility were elevated in prepubertal hCGαß+ mice, whereas Kiss1 expression was decreased prepubertally and Gad67 expression was elevated neonatally. The effect of androgens on the developmental programming of the hypothalamic-pituitary axis of hCGαß+ males was evaluated by perinatal and prepubertal antiandrogen (flutamide) administration. Our studies identified a critical window between gestational day 18 and postnatal day 14, during which chronically elevated androgens and/or their locally produced metabolites activate the hypothalamus and concomitantly shut-down the gonadotropin axis.


Asunto(s)
Andrógenos/metabolismo , Gonadotropina Coriónica Humana de Subunidad beta/metabolismo , Hormonas Glicoproteicas de Subunidad alfa/metabolismo , Sistema Hipotálamo-Hipofisario/crecimiento & desarrollo , Sistema Hipotálamo-Hipofisario/metabolismo , Antagonistas de Andrógenos/metabolismo , Animales , Aromatasa/genética , Aromatasa/metabolismo , Castración , Gonadotropina Coriónica Humana de Subunidad beta/genética , Hormona Folículo Estimulante/sangre , Hormona Folículo Estimulante/genética , Expresión Génica , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/metabolismo , Hormonas Glicoproteicas de Subunidad alfa/genética , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Humanos , Hipotálamo/fisiología , Kisspeptinas , Hormona Luteinizante/sangre , Hormona Luteinizante/genética , Masculino , Ratones , Ratones Transgénicos , Hipófisis/fisiología , Proteínas/genética , Proteínas/metabolismo , Pubertad/fisiología
20.
Endocr Relat Cancer ; 17(3): 611-21, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20453081

RESUMEN

The etiology of pituitary adenomas remains largely unknown, with the exception of involvement of estrogens in the formation of prolactinomas. We have examined the molecular pathogenesis of prolactin-producing pituitary adenomas in transgenic female mice expressing the human choriongonadotropin (hCG) beta-subunit. The LH/CG bioactivity is elevated in the mice, with consequent highly stimulated ovarian progesterone (P(4)) production, in the face of normal estrogen secretion. Curiously, despite normal estrogen levels, large prolactinomas developed in these mice, and we provide here several lines of evidence that the elevated P(4) levels are involved in the growth of these estrogen-dependent tumors. The antiprogestin mifepristone inhibited tumor growth, and combined postgonadectomy estradiol/P(4) treatment was more effective than estrogen alone in inducing tumor growth. Evidence for direct growth-promoting effect of P(4) was obtained from cultures of primary mouse pituitary cells and rat somatomammotroph GH3 cells. The mouse tumors and cultured cells revealed stimulation of the cyclin D1/cyclin-dependent kinase 4/retinoblastoma protein/transcription factor E2F1 pathway in the growth response to P(4). If extrapolated to humans, and given the importance of endogenous P(4) and synthetic progestins in female reproductive functions and their pharmacotherapy, it is relevant to revisit the potential role of these hormones in the origin and growth of prolactinomas.


Asunto(s)
Gonadotropina Coriónica Humana de Subunidad beta/metabolismo , Hormona Luteinizante/metabolismo , Neoplasias Hipofisarias/etiología , Progesterona/sangre , Prolactinoma/etiología , Análisis de Varianza , Animales , Proliferación Celular , Células Cultivadas , Gonadotropina Coriónica Humana de Subunidad beta/genética , Quinasa 4 Dependiente de la Ciclina/metabolismo , Femenino , Ratones , Ratones Transgénicos , Hipófisis/citología , Hipófisis/metabolismo , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/metabolismo , Prolactina/sangre , Prolactinoma/genética , Prolactinoma/metabolismo , Radioinmunoensayo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA