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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Gen Comp Endocrinol ; 336: 114247, 2023 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-36858273

RESUMO

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.


Assuntos
Gonadotropina Coriônica , Infertilidade , Humanos , Camundongos , Feminino , Animais , Gonadotropina Coriônica/farmacologia , Gonadotropina Coriônica Humana Subunidade beta/genética , Gonadotropina Coriônica Humana Subunidade beta/metabolismo , Camundongos Transgênicos , Luteinização , Mamíferos/metabolismo
2.
Reproduction ; 161(5): 539-548, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33730689

RESUMO

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.


Assuntos
Blastômeros/fisiologia , Fusão Celular , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário , Fertilização in vitro/veterinária , Tetraploidia , Animais , Blastômeros/citologia , Gatos , Transferência Embrionária , Embrião de Mamíferos/metabolismo , Feminino , Masculino , Panthera , Gravidez
3.
Front Neuroendocrinol ; 50: 118-122, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29074127

RESUMO

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.


Assuntos
Neoplasias Hipofisárias/metabolismo , Prolactinoma/metabolismo , Caracteres Sexuais , Fator de Crescimento Transformador beta1/metabolismo , Animais , Feminino , Humanos , Masculino
4.
Reproduction ; 148(4): R63-70, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25030890

RESUMO

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.


Assuntos
Gonadotropinas/metabolismo , Infertilidade Masculina/metabolismo , Reprodução , Animais , Hormônio Foliculoestimulante/genética , Hormônio Foliculoestimulante/metabolismo , Genótipo , Gonadotropinas/genética , Infertilidade Masculina/genética , Infertilidade Masculina/fisiopatologia , Hormônio Luteinizante/genética , Hormônio Luteinizante/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Fenótipo , Receptores do FSH/genética , Receptores do FSH/metabolismo , Receptores do LH/genética , Receptores do LH/metabolismo , Reprodução/genética , Transdução de Sinais
5.
Mol Cell Endocrinol ; 587: 112214, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38537882

RESUMO

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.


Assuntos
Neoplasias Ovarianas , Teratoma , Camundongos , Animais , Masculino , Feminino , Humanos , Lactente , Camundongos Transgênicos , Gonadotropina Coriônica/farmacologia , Oócitos , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia
6.
Neuroendocrinology ; 98(3): 212-23, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24080944

RESUMO

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.


Assuntos
Glutamato Descarboxilase/deficiência , Hormônio Liberador de Gonadotropina/deficiência , Hipotálamo Médio/metabolismo , Kisspeptinas/deficiência , Precursores de Proteínas/deficiência , Receptores de GABA-B/deficiência , Diferenciação Sexual/genética , Animais , Animais Recém-Nascidos , Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Núcleo Arqueado do Hipotálamo/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Glutamato Descarboxilase/genética , Hormônio Liberador de Gonadotropina/genética , Hipotálamo Médio/crescimento & desenvolvimento , Kisspeptinas/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Precursores de Proteínas/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores de GABA-B/genética
7.
Endocr Relat Cancer ; 29(6): 359-373, 2022 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-35324456

RESUMO

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.


Assuntos
Lactotrofos , Neoplasias Hipofisárias , Prolactinoma , Ativinas/metabolismo , Adulto , Animais , Feminino , Humanos , Hiperplasia , Inibinas/metabolismo , Inibinas/uso terapêutico , Lactotrofos/metabolismo , Lactotrofos/patologia , Camundongos , Ovariectomia , Neoplasias Hipofisárias/metabolismo , Prolactina/metabolismo , Prolactinoma/metabolismo , Prolactinoma/prevenção & controle
8.
Rev Endocr Metab Disord ; 12(4): 245-58, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21484328

RESUMO

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.


Assuntos
Modelos Animais de Doenças , Doenças do Sistema Endócrino/genética , Gonadotropinas/genética , Gonadotropinas/fisiologia , Camundongos Transgênicos , Animais , Doenças do Sistema Endócrino/metabolismo , Doenças do Sistema Endócrino/patologia , Doenças do Sistema Endócrino/fisiopatologia , Feminino , Gonadotropinas/deficiência , Gonadotropinas/metabolismo , Humanos , Masculino , Camundongos , Ovário/metabolismo , Ovário/patologia , Ovário/fisiologia , Hipófise/metabolismo , Hipófise/patologia , Hipófise/fisiologia , Testículo/metabolismo , Testículo/patologia , Testículo/fisiologia
9.
Artigo em Inglês | MEDLINE | ID: mdl-30761084

RESUMO

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.

10.
Endocr Relat Cancer ; 26(5): 497-510, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30856609

RESUMO

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.


Assuntos
Neoplasias Hipofisárias/prevenção & controle , Progesterona/farmacologia , Prolactina/metabolismo , Prolactinoma/prevenção & controle , Receptores de Dopamina D2/fisiologia , Receptores de Progesterona/agonistas , Animais , Gonadotropina Coriônica Humana Subunidade beta/genética , Gonadotropina Coriônica Humana Subunidade beta/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Neoplasias Hipofisárias/etiologia , Neoplasias Hipofisárias/patologia , Prolactinoma/etiologia , Prolactinoma/patologia , Ratos , Transdução de Sinais
12.
J Endocrinol ; 232(3): 535-546, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28096433

RESUMO

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.


Assuntos
Gonadotropina Coriônica Humana Subunidade beta/metabolismo , Hipófise/metabolismo , Neoplasias Hipofisárias/metabolismo , Prolactinoma/metabolismo , Caracteres Sexuais , Fator de Crescimento Transformador beta1/metabolismo , Animais , Gonadotropina Coriônica Humana Subunidade beta/genética , Feminino , Proteínas de Ligação a TGF-beta Latente/genética , Proteínas de Ligação a TGF-beta Latente/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Hipófise/patologia , Neoplasias Hipofisárias/genética , Neoplasias Hipofisárias/patologia , Prolactinoma/genética , Prolactinoma/patologia , Proteína Smad4/genética , Proteína Smad4/metabolismo , Proteína Smad7/genética , Proteína Smad7/metabolismo
13.
Oncogene ; 24(49): 7301-9, 2005 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-16007123

RESUMO

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.


Assuntos
Adenoma/etiologia , Gonadotropina Coriônica Humana Subunidade beta/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Subunidade alfa de Hormônios Glicoproteicos/metabolismo , Células Intersticiais do Testículo/metabolismo , 3-Hidroxiesteroide Desidrogenases/metabolismo , Adenoma/metabolismo , Adenoma/patologia , Animais , Gonadotropina Coriônica Humana Subunidade beta/genética , Feto , Regulação Neoplásica da Expressão Gênica , Subunidade alfa de Hormônios Glicoproteicos/genética , Humanos , Oxirredutases Intramoleculares/metabolismo , Células Intersticiais do Testículo/citologia , Lipocalinas , Masculino , Camundongos , Camundongos Transgênicos , Fenótipo , Testículo/metabolismo , Testículo/patologia , Trombospondinas/metabolismo , Regulação para Cima
14.
Mol Cell Endocrinol ; 252(1-2): 126-35, 2006 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-16675102

RESUMO

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+).


Assuntos
Animais Geneticamente Modificados , Hormônio Luteinizante/fisiologia , Camundongos/genética , Animais , Feminino , Masculino , Camundongos Knockout , Camundongos Transgênicos , Ovário/fisiologia , Receptores do LH/deficiência , Receptores do LH/genética , Testículo/fisiologia
15.
J Endocrinol ; 230(1): 157-69, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27154336

RESUMO

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.


Assuntos
Gonadotropina Coriônica Humana Subunidade beta/metabolismo , Intolerância à Glucose/metabolismo , Hiperinsulinismo/metabolismo , Hiperprolactinemia/metabolismo , Hipertrigliceridemia/metabolismo , Resistência à Insulina/fisiologia , Animais , Glicemia/metabolismo , Cabergolina , Gonadotropina Coriônica Humana Subunidade beta/genética , Ergolinas/uso terapêutico , Feminino , Intolerância à Glucose/tratamento farmacológico , Intolerância à Glucose/genética , Hiperinsulinismo/tratamento farmacológico , Hiperinsulinismo/genética , Hiperprolactinemia/tratamento farmacológico , Hiperprolactinemia/genética , Hipertrigliceridemia/tratamento farmacológico , Hipertrigliceridemia/genética , Insulina/sangue , Camundongos , Camundongos Transgênicos , Prolactina/sangue , Triglicerídeos/sangue
16.
Expert Opin Biol Ther ; 15(8): 1183-90, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26160491

RESUMO

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.


Assuntos
Gonadotropina Coriônica/antagonistas & inibidores , Descoberta de Drogas/tendências , Vacinas Anticoncepcionais/administração & dosagem , Animais , Antineoplásicos Hormonais/administração & dosagem , Gonadotropina Coriônica/química , Gonadotropina Coriônica/imunologia , Feminino , Humanos , Masculino , Gravidez , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/imunologia , Vacinação/métodos , Vacinas Anticoncepcionais/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia
17.
Endocrinology ; 144(11): 4980-90, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12960071

RESUMO

We previously developed a transgenic (TG) mouse model that overexpresses the human chorionic gonadotropin (hCG) beta-subunit under the universal human ubiquitin C promoter, displaying in males a modest 3-fold increase in circulating levels of LH/hCG bioactivity. The males were fertile and presented with a mild reproductive phenotype. To achieve higher levels of hCG, a double TG model was generated by cross-breeding the hCG beta-expressing mice with another TG line harboring a ubiquitin C/common alpha-subunit fusion gene. The double-TG mice expressed excessive levels of dimeric hCG, with 2000-fold elevated circulating LH/hCG bioactivity. These male mice were infertile, primarily due to inability to copulate, and they showed enhanced testicular androgen production despite clear down-regulation of LH/hCG receptors. Their intratesticular inhibin B was unaltered, but serum FSH was markedly reduced. Apparently the chronic hCG hyperstimulation led to focal Leydig cell proliferation/hypertrophy at 6 months of age, but failed to promote testicular tumors. Even though full spermatogenesis occurred in most of the seminiferous tubules, progressive tubule degeneration was apparent as the males grew older. The prostate and seminal vesicles were enlarged by distension of glandular lumina. Functional urethral obstruction was indicated by distension and sperm accumulation in distal vas deferens as well as by dilated urinary bladder and enlarged kidneys. The abnormal function of accessory sex glands and/or lower urinary tract as a consequence of the disturbed sex hormone balance or direct action of hCG may be the main cause of infertility in this model. The present study provides in vivo evidence that exposure of male mice to chronically elevated levels of hCG severely affects their urogenital tract function at multiple sites and causes infertility, but, unlike in LH/hCG overexpressing female mice, it is not tumorigenic.


Assuntos
Gonadotropina Coriônica Humana Subunidade beta/metabolismo , Genitália Masculina/patologia , Genitália Masculina/fisiopatologia , Subunidade alfa de Hormônios Glicoproteicos/metabolismo , Esteroides/biossíntese , Animais , Peso Corporal , Linhagem Celular , Expressão Gênica , Hormônios/sangue , Hormônios/metabolismo , Infertilidade Masculina/etiologia , Masculino , Camundongos , Camundongos Transgênicos , Tamanho do Órgão , Receptores do LH/metabolismo , Testículo/metabolismo , Transgenes , Sistema Urinário/patologia
18.
Endocrinology ; 143(10): 4084-95, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12239120

RESUMO

To assess the consequences of prolonged exposure to elevated levels of LH/human chorionic gonadotropin (hCG) in the female, we developed a transgenic (TG) mouse model (hCGbeta+) that overexpresses the hCGbeta-subunit cDNA. Because of the promoter used, ubiquitin C, the transgene is expressed in multiple tissues, including the pituitary gland, in which coupling with the endogenous common alpha-subunit results in synthesis of high levels of bioactive hCG. The TG females presented with precocious puberty, infertility, enhanced ovarian steroidogenesis, and abnormal uterine structure. Pituitary enlargement was evident from the age of 2 months, which progressed to adenomas by the age of 10-12 months. Immunohistochemical studies and electron microscopy demonstrated lactotrope origin for the adenomas, associated with severe hyperprolactinemia. The mammary glands of TG females showed marked lobuloalveolar development followed by mammary tumors with characteristics of adenocarcinoma at the age of 9-12 months. More than 90% of penetrance and high frequency of metastasis (47%) was observed. Formation of the pituitary and mammary gland tumors was totally abolished by ovariectomy despite persistently elevated hCG levels. Taken together, these findings suggest that the hCG-induced aberrations of ovarian function are clearly responsible for the extragonadal tumors observed in these TG mice.


Assuntos
Adenoma/induzido quimicamente , Gonadotropina Coriônica Humana Subunidade beta , Infertilidade/induzido quimicamente , Neoplasias Mamárias Experimentais/induzido quimicamente , Adeno-Hipófise , Neoplasias Hipofisárias/induzido quimicamente , Prolactina/metabolismo , Adenoma/metabolismo , Adenoma/patologia , Animais , Gonadotropina Coriônica Humana Subunidade beta/administração & dosagem , Gonadotropina Coriônica Humana Subunidade beta/genética , Relação Dose-Resposta a Droga , Feminino , Genitália Feminina/patologia , Genitália Feminina/fisiopatologia , Hormônios/biossíntese , Humanos , Hiperprolactinemia/induzido quimicamente , Infertilidade/patologia , Infertilidade/fisiopatologia , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Transgênicos/genética , Obesidade/induzido quimicamente , Ovário/metabolismo , Adeno-Hipófise/patologia , Neoplasias Hipofisárias/metabolismo , Neoplasias Hipofisárias/patologia
19.
Reprod Biol ; 14(1): 9-15, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24607250

RESUMO

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.


Assuntos
Gonadotropina Coriônica/metabolismo , Hiperprolactinemia/metabolismo , Receptores do LH/metabolismo , Animais , Gonadotropina Coriônica/genética , Modelos Animais de Doenças , Feminino , Hiperprolactinemia/genética , Camundongos , Camundongos Transgênicos , Hipófise/metabolismo , Receptores do LH/genética
20.
Mol Cell Endocrinol ; 390(1-2): 102-11, 2014 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-24755422

RESUMO

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.


Assuntos
Gonadotropina Coriônica/metabolismo , Hipotálamo/metabolismo , Puberdade Precoce/metabolismo , Antagonistas de Androgênios/farmacologia , Antagonistas de Androgênios/uso terapêutico , Animais , Aromatase/metabolismo , Células Cultivadas , Gonadotropina Coriônica/fisiologia , Estradiol/sangue , Feminino , Flutamida/farmacologia , Flutamida/uso terapêutico , Hormônio Foliculoestimulante/sangue , Expressão Gênica , Hormônio Liberador de Gonadotropina/fisiologia , Humanos , Camundongos Transgênicos , Hipófise/metabolismo , Puberdade Precoce/tratamento farmacológico , Testosterona/sangue , Vagina/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA