Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
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
2.
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
3.
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
4.
Reproduction ; 156(4): R101-R109, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30304933

RESUMEN

In mammals, the reproductive function is controlled by the hypothalamic­pituitary­gonadal axis. During development, mechanisms mediated by gonadal steroids exert an imprinting at the hypothalamic­pituitary level, by establishing sexual differences in the circuits that control male and female reproduction. In rodents, the testicular production of androgens increases drastically during the fetal/neonatal stage. This process is essential for the masculinization of the reproductive tract, genitals and brain. The conversion of androgens to estrogens in the brain is crucial for the male sexual differentiation and behavior. Conversely, feminization of the brain occurs in the absence of high levels of gonadal steroids during the perinatal period in females. Potential genetic contribution to the differentiation of brain cells through direct effects of genes located on sex chromosomes is also relevant. In this review, we will focus on the phenotypic alterations that occur on the hypothalamic­pituitary­gonadal axis of transgenic mice with persistently elevated expression of the human chorionic gonadotropin hormone (hCG). Excess of endogenously synthesized gonadal steroids due to a constant hCG stimulation is able to disrupt the developmental programming of the hypothalamic­pituitary axis in both transgenic males and females. Locally produced estrogens by the hypothalamic aromatase might play a key role in the phenotype of these mice. The 'four core genotypes' mouse model demonstrated a potential influence of sex chromosome genes in brain masculinization before critical periods of sex differentiation. Thus, hormonal and genetic factors interact to regulate the local production of the neurosteroids necessary for the programming of the male and female reproductive function.


Asunto(s)
Hormonas Esteroides Gonadales/fisiología , Sistema Hipotálamo-Hipofisario/fisiología , Desarrollo Sexual , Animales , Ratones , Ratones Transgénicos
5.
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
6.
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
7.
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
8.
Endocrinology ; 163(9)2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35863039

RESUMEN

Sex differences in the control of prolactin secretion are well documented. Sex-related differences in intrapituitary factors regulating lactotroph function have recently attracted attention. Sex differences in prolactinoma development are well documented in clinic, prolactinomas being more frequent in women but more aggressive in men, for poorly understood reasons. Kallikrein, the enzyme releasing kinins has been found in the pituitary, but there is no information on pituitary kinin receptors and their function. In the present work, we characterized pituitary bradykinin receptors (BRs) at the messenger RNA and protein levels in 2 mouse models of prolactinoma, Drd2 receptor gene inactivation and hCGß gene overexpression, in both males and females, wild type or genomically altered. BR B2 (B2R) accounted for 97% or more of total pituitary BRs in both models, regardless of genotype, and was present in lactotrophs, somatotrophs, and gonadotrophs. Male pituitaries displayed higher level of B2R than females, regardless of genotype. Pituitary B2R gene expression was downregulated by estrogen in both males and females but only in females by dopamine. Activation of B1R or B2R by selective pharmacological agonists induced prolactin release in male pituitaries but inhibited prolactin secretion in female pituitaries. Increased B2R content was observed in pituitaries of mutated animals developing prolactinomas, compared to their respective wild-type controls. The present study documents a novel sex-related difference in the control of prolactin secretion and suggests that kinins are involved, through B2R activation, in lactotroph function and prolactinoma development.


Asunto(s)
Neoplasias Hipofisarias , Prolactinoma , Animales , Femenino , Humanos , Cininas , Masculino , Ratones , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/metabolismo , Prolactina/metabolismo , Prolactinoma/genética , Prolactinoma/metabolismo , Receptor de Bradiquinina B2/agonistas , Receptor de Bradiquinina B2/genética , Receptor de Bradiquinina B2/metabolismo , Receptores de Bradiquinina
9.
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
10.
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
11.
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.

12.
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
14.
Endocrinology ; 148(8): 3694-703, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17510243

RESUMEN

Transgenic (TG) mice expressing human chorionic gonadotropin (hCG) beta-subunit under the ubiquitin C promoter, presenting with a moderately elevated level of LH/hCG bioactivity develop multiple neoplasms secondary to the endocrine abnormalities, including mammary gland tumors after the age of 9 months. The increased levels of circulating estradiol, progesterone, and prolactin of the TG females after puberty boost the lobuloalveolar development in the mammary gland resulting ultimately in the formation of estrogen and progesterone receptor-negative, malignant tumors. These tumors have a similar histopathology with those observed in TG mice with activated wnt/beta-catenin pathway, showing increased expression of beta-catenin, also a common finding in human breast tumors. Transdifferentiation is observed in mammary tumors of the hCGbeta TG mice, accompanied by abnormal expression of the Wnt genes in the tumorous and nontumorous mammary gland tissue. Specifically we found increased expression of Wnt5b in the TG mammary glands at the age of 3 months and up-regulation of Wnt7b and -5b in the subsequently appearing tumors. Importantly, hCG was found to up-regulate these wnt ligands in mouse mammary gland, independent of the changes in ovarian steroidogenesis. Thus, the hCGbeta-overexpressing TG mice represent a novel model that links enhanced hCG action to dysregulated wnt signaling in the mammary gland, resulting in beta-catenin-stabilizing mammary tumorigenesis. The novel finding of hCG up-regulating wnt7b and wnt5b could contribute to pregnancy-induced breast cancer in humans.


Asunto(s)
Gonadotropina Coriónica Humana de Subunidad beta/genética , Gonadotropina Coriónica Humana de Subunidad beta/metabolismo , Glicoproteínas/metabolismo , Neoplasias Mamarias Animales/fisiopatología , Mioepitelioma/fisiopatología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Wnt/metabolismo , Factores de Edad , Animales , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Glándulas Mamarias Animales/patología , Glándulas Mamarias Animales/fisiología , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Ratones , Ratones Endogámicos , Ratones Transgénicos , Mioepitelioma/metabolismo , Mioepitelioma/patología , Ovariectomía , Receptores de Progesterona/metabolismo , Regulación hacia Arriba/fisiología , beta Catenina/metabolismo
15.
Mol Cell Endocrinol ; 260-262: 255-63, 2007 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-17029767

RESUMEN

In order to study the physiology and pathophysiology of gonadotrophin action, we have produced transgenic (TG) mice overexpressing human chorionic gonadotrophin (hCG) alpha and beta subunits (hCG+ mice) and knockout (KO) mice for the luteinising hormone receptor (LHR; LuRKO mice). The two extremes in LH function, i.e. strong LH/hCG stimulation and total blockade of this action, confirm numerous earlier concepts about LH function, but they also reveal new aspects about gonadal function during excessive LH production and in the absence of this trophic stimulus. The purpose of this review is to summarise the key findings on these two genetically modified mouse models.


Asunto(s)
Gonadotropina Coriónica/deficiencia , Gonadotropina Coriónica/genética , Hormona Luteinizante/deficiencia , Hormona Luteinizante/genética , Animales , Expresión Génica , Genitales/citología , Humanos , Ratones , Ratones Noqueados , Fenotipo
16.
Mol Cell Endocrinol ; 269(1-2): 9-16, 2007 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-17350753

RESUMEN

Gonadotropins are indispensable in both sexes in the regulation of gonadal sex steroid production and gametogenesis. In addition to their well-established classical actions, numerous recent publications have indicated the presence and function of luteinizing hormone/chorionic gonadotropin receptors (LH/hCG-R) in a variety of extragonadal tissues. However, the physiological significance of such effects has remained unclear. We have generated two genetically modified mouse models, one with excessive production of hCG and the other with targeted disruption of LH/hCG-R gene, and used them to address the functions of LH and hCG. Numerous gonadal and extragonadal phenotypes were found in the models with the two extremes of LH/hCG action. However, when the extragonadal effects were scrutinized in greater detail, they all appeared to arise through modification of gonadal function, either through enhanced or inhibited response to LH/hCG stimulation. Hence, further evidence is needed before the extragonadal LH/hCG-R expression can be considered functionally significant.


Asunto(s)
Gonadotropinas/fisiología , Animales , Huesos/efectos de los fármacos , Femenino , Gonadotropinas/biosíntesis , Gonadotropinas/genética , Gonadotropinas/farmacología , Gónadas/efectos de los fármacos , Gónadas/metabolismo , Humanos , Ratones , Ratones Transgénicos , Ovario/trasplante
17.
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
18.
Oncogene ; 24(49): 7301-9, 2005 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-16007123

RESUMEN

We have previously demonstrated that male transgenic (TG) mice overexpressing human chorionic gonadotropin (hCG+) develop reproductive organ defects, but no tumors, in adult age. In this study, the effects of persistently elevated hCG were followed in TG males between day 5 postpartum and adulthood. Leydig cell (LC) adenomas were found in prepubertal mice, most prominently at the age of 10 days, but not in adult age. Serum testosterone concentrations were significantly increased in TG males at all ages studied. The phenotype of the prepubertal hCG+ males resembled that found in boys upon expression of constitutively activating luteinizing hormone (LH) receptor mutations. The temporal expression patterns of the fetal LC marker gene, thrombospondin 2, and those of adult LCs, hydroxysteroid dehydrogenase-6, delta5-3-beta and prostaglandin D synthase, were similar in wild-type and hCG+ males. Hence, the postnatal adenomas resemble functionally fetal LCs, and only these cells are susceptible to hCG-induced tumorigenesis. Our findings demonstrate a novel intriguing difference between the fetal and adult LC populations and provide further insight into the potential tumorigenic effects of gonadotropins.


Asunto(s)
Adenoma/etiología , Gonadotropina Coriónica Humana de Subunidad beta/metabolismo , Regulación del Desarrollo de la Expresión Génica , Hormonas Glicoproteicas de Subunidad alfa/metabolismo , Células Intersticiales del Testículo/metabolismo , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , Adenoma/metabolismo , Adenoma/patología , Animales , Gonadotropina Coriónica Humana de Subunidad beta/genética , Feto , Regulación Neoplásica de la Expresión Génica , Hormonas Glicoproteicas de Subunidad alfa/genética , Humanos , Oxidorreductasas Intramoleculares/metabolismo , Células Intersticiales del Testículo/citología , Lipocalinas , Masculino , Ratones , Ratones Transgénicos , Fenotipo , Testículo/metabolismo , Testículo/patología , Trombospondinas/metabolismo , Regulación hacia Arriba
19.
Mol Cell Endocrinol ; 252(1-2): 126-35, 2006 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-16675102

RESUMEN

Numerous genetically modified mouse models have recently been developed for the study of the pituitary-gonadal interactions. They include spontaneous or engineered knockouts (KO) of the gonadotrophin-releasing hormone (GnRH) and its receptor, the gonadotrophin common-alpha(Calpha), luteinising hormone (LH) beta and follicle-stimulating hormone (FSH) beta subunits, and the two gonadotrophin receptors (R), LHR and FSHR. In addition, there are also transgenic (TG) mice overexpressing gonadotrophin subunits and producing supraphysiological levels of these hormones. These models have offered relevant phenocopies for similar mutations in humans and to a great extent expanded our knowledge on normal and pathological functions of the hypothalamic-pituitary-gonadal (HPG) axis. The purpose of this article is to review some of our recent findings on two such mouse models, the LHR KO mouse (LuRKO), and the hCG overexpressing TG mouse (hCG+).


Asunto(s)
Animales Modificados Genéticamente , Hormona Luteinizante/fisiología , Ratones/genética , Animales , Femenino , Masculino , Ratones Noqueados , Ratones Transgénicos , Ovario/fisiología , Receptores de HL/deficiencia , Receptores de HL/genética , Testículo/fisiología
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA