Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 154
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 111(34): 12492-7, 2014 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-25114230

RESUMEN

Oocyte endowment dwindles away during prepubertal and adult life until menopause occurs, and apoptosis has been identified as a central mechanism responsible for oocyte elimination. A few recent reports suggest that uncontrolled inflammation may adversely affect ovarian reserve. We tested the possible role of the proinflammatory cytokine IL-1 in the age-related exhaustion of ovarian reserve using IL-1α and IL-1ß-KO mice. IL-1α-KO mice showed a substantially higher pregnancy rate and litter size compared with WT mice at advanced age. The number of secondary and antral follicles was significantly higher in 2.5-mo-old IL-1α-KO ovaries compared with WT ovaries. Serum anti-Müllerian hormone, a putative marker of ovarian reserve, was markedly higher in IL-1α-KO mice from 2.5 mo onward, along with a greater ovarian response to gonadotropins. IL-1ß-KO mice displayed a comparable but more subtle prolongation of ovarian lifespan compared with IL-1α-KO mice. The protein and mRNA of both IL-1α and IL-1ß mice were localized within the developing follicles (oocytes and granulosa cells), and their ovarian mRNA levels increased with age. Molecular analysis revealed decreased apoptotic signaling [higher B-cell lymphoma 2 (BCL-2) and lower BCL-2-associated X protein levels], along with a marked attenuation in the expression of genes coding for the proinflammatory cytokines IL-1ß, IL-6, and TNF-α in ovaries of IL-1α-KO mice compared with WT mice. Taken together, IL-1 emerges as an important participant in the age-related exhaustion of ovarian reserve in mice, possibly by enhancing the expression of inflammatory genes and promoting apoptotic pathways.


Asunto(s)
Interleucina-1alfa/deficiencia , Interleucina-1beta/deficiencia , Ovario/fisiología , Envejecimiento , Animales , Hormona Antimülleriana/sangre , Apoptosis , Femenino , Expresión Génica , Mediadores de Inflamación/metabolismo , Interleucina-1alfa/genética , Interleucina-1alfa/fisiología , Interleucina-1beta/genética , Interleucina-1beta/fisiología , Tamaño de la Camada , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ovario/citología , Ovario/inmunología , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de HFE/genética , Receptores de HFE/fisiología , Receptores Tipo I de Interleucina-1/deficiencia , Receptores Tipo I de Interleucina-1/genética , Receptores Tipo I de Interleucina-1/fisiología
2.
Reprod Domest Anim ; 52 Suppl 2: 41-44, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28402061

RESUMEN

This study aimed to evaluate and compare the ovarian and uterine characteristics along with the ovarian mRNA and protein expression of LHR and FSHR between the pre-pubertal and adult female cats. The uterine horns and ovaries were collected from pre-pubertal and adult female cats at their follicular, luteal and interoestrous stages of the oestrous cycle (n = 6/group). Endometrial and myometrial thickness, uterine gland diameter, ovarian weight and type of follicles were analysed. The mRNA and protein expression of LHR and FSHR was analysed by IHC and qPCR, respectively. The ovarian weight of pre-pubertal cats was significantly lower than that of adult cats. No differences were recorded in the numbers of primordial and primary follicles between the study groups, while adult luteal cats had significantly lower numbers of antral follicles compared to pre-pubertal cats. No differences in the ovarian expression of FSHR mRNA, LHR protein or mRNA were found between the pre-pubertal and adult cats, but significantly lower FSHR protein expression was found in pre-pubertal cats compared to adult luteal cats.


Asunto(s)
Folículo Ovárico/fisiología , Receptores de HFE/fisiología , Receptores de HL/fisiología , Útero/fisiología , Animales , Gatos , Ciclo Estral/fisiología , Femenino , Expresión Génica
3.
Artículo en Inglés | MEDLINE | ID: mdl-27164487

RESUMEN

Previous works on European sea bass have determined that long-term exposure to restrictive feeding diets alters the rhythms of some reproductive/metabolic hormones, delaying maturation and increasing apoptosis during gametogenesis. However, exactly how these diets affect key genes and hormones on the brain-pituitary-gonad (BPG) axis to trigger puberty is still largely unknown. We may hypothesize that all these signals could be integrated, at least in part, by the kisspeptin system. In order to capture a glimpse of these regulatory mechanisms, kiss1 and kiss2 mRNA expression levels and those of their kiss receptors (kiss1r, kiss2r) were analyzed in different areas of the brain and in the pituitary of pubertal male sea bass during gametogenesis. Furthermore, other reproductive hormones and factors as well as the percentage of males showing full spermiation were also analyzed. Treated fish fed maintenance diets provided evidence of overexpression of the kisspeptin system in the main hypophysiotropic regions of the brain throughout the entire sexual cycle. Conversely, Gnrh1 and gonadotropin pituitary content and plasma sexual steroid levels were downregulated, except for Fsh levels, which were shown to increase during spermiation. Treated fish exhibited lower rates of spermiation as compared to control group and a delay in its accomplishment. These results demonstrate how the kisspeptin system and plasma Fsh levels are differentially affected by maintenance diets, causing a retardation, but not a full blockage of the reproductive process in the teleost fish European sea bass. This suggests that a hormonal adaptive strategy may be operating in order to preserve reproductive function in this species.


Asunto(s)
Lubina/fisiología , Proteínas de Peces/fisiología , Alimentos , Kisspeptinas/fisiología , Reproducción/fisiología , Maduración Sexual/fisiología , Animales , Lubina/genética , Proteínas de Peces/genética , Hormona Folículo Estimulante/sangre , Hormona Folículo Estimulante/metabolismo , Expresión Génica , Hormona Liberadora de Gonadotropina/metabolismo , Gonadotropinas/sangre , Gonadotropinas/metabolismo , Hipotálamo/metabolismo , Kisspeptinas/genética , Hormona Luteinizante/metabolismo , Masculino , Mesencéfalo/metabolismo , Hipófisis/metabolismo , Prosencéfalo/metabolismo , Receptores de HFE/genética , Receptores de HFE/fisiología , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/fisiología , Receptores de HL/genética , Receptores de HL/fisiología , Reproducción/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estaciones del Año , Maduración Sexual/genética , Espermatogénesis/genética , Espermatogénesis/fisiología
4.
Biochem Biophys Res Commun ; 461(3): 525-32, 2015 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-25911324

RESUMEN

Our previous study showed that Octamer-binding transcription factor 4 (OCT4) expression was upregulated and significantly associated with histological grade through the analysis of OCT4 expression in 159 ovarian cancer tissue samples, and OCT4 mediated follicle-stimulating hormone (FSH)-induced anti-apoptosis in epithelial ovarian cancer. Nevertheless, whether OCT4 participates in FSH-induced invasion in ovarian cancer is still unknown. Therefore, the present study aimed to define whether FSH-induced ovarian cancer invasion is mediated by OCT4. In present study, we showed that FSH induced not only the epithelial-mesenchymal transition (EMT) and invasive phenotype but also the upregulation of OCT4 expression in a dose- and time-dependent manner in epithelial ovarian cancer cells. In addition, the expression of FSH receptor (FSHR) was upregulated by FSH induction, and knockdown of FSHR inhibited FSH-stimulated OCT4 expression. ERK1/2 signaling pathway participated in the enhanced expression of OCT4 and Snail induced by FSH. We further showed that the activated expression of Snail and N-cadherin, the suppressed expression of E-cadherin and the morphological change of the cells stimulated by FSH were blocked by OCT4-specific small interfering RNA. Moreover, our results showed that OCT4 mediated the increase in invasive capacity induced by FSH in ovarian cancer cells. Taken together, our work reveals that OCT4 is an essential mediator in FSH-induced EMT and invasion in epithelial ovarian cancer and may act as a potential therapeutic target.


Asunto(s)
Hormona Folículo Estimulante/fisiología , Sistema de Señalización de MAP Quinasas , Invasividad Neoplásica , Neoplasias Glandulares y Epiteliales/patología , Factor 3 de Transcripción de Unión a Octámeros/fisiología , Neoplasias Ováricas/patología , Secuencia de Bases , Carcinoma Epitelial de Ovario , Humanos , Neoplasias Glandulares y Epiteliales/enzimología , Neoplasias Glandulares y Epiteliales/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/metabolismo , Reacción en Cadena de la Polimerasa , ARN Interferente Pequeño , Receptores de HFE/fisiología
5.
Clin Calcium ; 24(6): 815-9, 2014 Jun.
Artículo en Japonés | MEDLINE | ID: mdl-24870831

RESUMEN

Pathophysiology of postmenopausal osteoporosis has been considered due to deficiency of estrogen. However, it has been reported that the rate of bone mass loss during perimenopause is greater than that in postmenopause, probably due to increased FSH. From the recent knowledge of basic research on FSH, FSH can directly stimulate osteoclast formation and accelerate bone resorption. In contrast, FSH transgenic mice exhibit increased bone mass dependent on ovarian function. In this review, the controversies on the function of FSH in bone mass regulation will be discussed.


Asunto(s)
Huesos/metabolismo , Hormona Folículo Estimulante/fisiología , Gonadotropinas/metabolismo , Animales , Densidad Ósea , Resorción Ósea , Estrógenos/deficiencia , Estrógenos/fisiología , Humanos , Osteoclastos , Osteoporosis Posmenopáusica/metabolismo , Ligando RANK , Receptores de HFE/fisiología
6.
J Biol Regul Homeost Agents ; 27(3): 805-16, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24152845

RESUMEN

Cadmium (Cd), an ubiquitous environmental metal, mainly used for industrial purposes, may be toxic at level of the reproductive system. Testis tubular-based Sertoli cells (SC), play a major role in constituting the blood-testis barrier and provide a unique microenvironment for the genesis and differentiation of germ cells. Hence SC strictly control sperm qualitative and quantitative parameters. We aimed to assess whether exposure to Cd would adversely affect superior mammal SC viability and function. We isolated and purified SC from pre-pubertal pig testes according to our method and incubated the retrieved cells with three different Cadmium chloride concentrations (5-10-15 microM). Parameters of SC function such as inhibin B and anti-Mullerian hormone (AMH) were depressed by Cd exposure, contrary to what observed in untreated controls. No impairment of the FSH receptor integrity on the SC, as assessed by 17-beta-estradiol production, upon stimulation with FSH, was observed in either 5 microM Cd-treated or untreated controls. Differences, on the contrary, were observed for higher Cd concentrations (10 and 15 mM), in terms of FSH receptor integrity, that was altered, as compared to untreated controls, in terms of lower production of 17-beta-estradiol. In addition, the apoptotic test showed a significant increase of early (ANNEXIN V-/Propidium Iodide+) (AV-/PI+) and late apoptotic cells (AV+/ PI+) in all Cd -treated SC conditions as compared to controls. In conclusion, the Cd -related toxicity on SC, clearly demonstrated by our study, even at low concentrations, is expected to damage spermatogenesis that directly is dependent upon retention of SC viability and function.


Asunto(s)
Cadmio/toxicidad , Células de Sertoli/efectos de los fármacos , Animales , Hormona Antimülleriana/metabolismo , Apoptosis/efectos de los fármacos , Cadmio/farmacocinética , Supervivencia Celular/efectos de los fármacos , Inhibinas/metabolismo , Masculino , Receptores de HFE/efectos de los fármacos , Receptores de HFE/fisiología , Células de Sertoli/fisiología , Porcinos
7.
Andrologia ; 45(5): 339-44, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23013557

RESUMEN

Follicle-stimulating hormone (FSH), interacting with its receptor (FSHR), participates in the production of spermatozoa and androgens. Androgens exert their effects on male sex determination, development and sperm production by binding to androgen receptor (AR). In the present study, we sought to explore the potential synergistic effects of FSHR and AR gene variants on sperm quality. 200 oligozoospermic and 250 normozoospermic men were examined. DNA was extracted from spermatozoa, and the FSHR 307 (T/A), FSHR 680 (N/S) and AR (CAG)n polymorphisms were genotyped. Their parallel analysis revealed six combined genotypes. A gradual reduction of sperm motility, from long AR allele-Thr307Thr/Asn680Asn carriers to long AR allele-Ala307Ala/Ser680Ser carriers and from short AR allele-Thr307Thr/Asn680Asn carriers to short AR allele-Ala307Ala/Ser680Ser carriers was revealed in normozoospermic men (P < 0.001). Similar associations were observed in oligozoospermic men (P < 0.001). In our series, the synergism of the long AR alleles with the FSHRThr307/Asn680 allelic variant was associated with increased sperm motility, while the synergism of the short AR alleles with the FSHRAla307/Ser680 allelic variant was associated with decreased motility, supporting the significance of these genes in semen quality.


Asunto(s)
Oligospermia/genética , Receptores Androgénicos/genética , Receptores de HFE/genética , Análisis de Semen , Adulto , Alelos , Genotipo , Humanos , Masculino , Polimorfismo Genético , Receptores Androgénicos/fisiología , Receptores de HFE/fisiología , Motilidad Espermática/genética
8.
J Assist Reprod Genet ; 30(5): 717-21, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23536150

RESUMEN

BACKGROUND: Polycystic ovary syndrome (PCOS) is a common complex genetic endocrinopathy. It has high heritability, and twin studies indicate that it is a complex polygenic disorder. Searching for major genes of PCOS is crucial to clarify its molecular pathogenesis. A previous genome-wide association study in Chinese women with PCOS identified a region on chromosome 2p16.3 that encodes the follicle-stimulating hormone receptor (FSHR) genes as a reproducible PCOS susceptibility locus. In the present study, we performed a replication analysis of the association between two common variants of the FSHR gene and PCOS in Northern Chinese Han women. RESULTS: We recruited 384 unrelated PCOS patients and 768 healthy individuals from the Shaanxi province in the northern part of China. Two polymorphisms (Ala307Thr and Ser680Asn) of the FSHR gene and the clinical characteristics of the study subjects were analyzed in the case-control sample. The frequency of FSHR Ala307Thr and Ser680Asn variants along with the haplotype was not significantly different between the PCOS patients and the controls; however, the Ser680 variants may be associated with high levels of FSH and low E2 levels. CONCLUSION: The variant of Ser680 was not associated with PCOS but it may be related to high FSH levels. The present study suggests that the two variants of the FSHR gene are not a causative factor of PCOS in Northern Chinese Han women.


Asunto(s)
Sustitución de Aminoácidos/fisiología , Pueblo Asiatico/genética , Síndrome del Ovario Poliquístico/genética , Receptores de HFE/genética , Adulto , Alanina/genética , Asparagina/genética , Estudios de Casos y Controles , China/epidemiología , Femenino , Frecuencia de los Genes/fisiología , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Geografía , Humanos , Mutación Missense/fisiología , Síndrome del Ovario Poliquístico/epidemiología , Síndrome del Ovario Poliquístico/etnología , Polimorfismo de Nucleótido Simple/fisiología , Receptores de HFE/fisiología , Serina/genética , Treonina/genética
9.
Biol Reprod ; 86(3): 77, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22116805

RESUMEN

The aryl hydrocarbon receptor (AHR) mediates the toxicity of a variety of environmental chemicals. Apart from this, an understanding is emerging that the AHR has a fundamental role in female reproduction. Evidence suggests that AHR participates in regulation of follicle-stimulating hormone receptor (Fshr) transcript level in mouse ovary by binding to the promoter of this gene in vivo. The purpose of this study was to demonstrate the molecular interplay of the Fshr promoter involved in the transactivation by AHR in mouse granulosa cells. We found that AHR activates the Fshr promoter through the region from -209 to -99 bp. In this region, the importance of the E-box motif was revealed by site-directed mutagenesis followed by promoter analysis. By focusing on the DNA/protein interactions, we defined the fact that the intact E-box but not upstream transcription factor 1 (USF1), which is known to bind this motif, is necessary for AHR binding to mouse Fshr promoter. Furthermore, by constructing AHR mutants defective in DNA interaction, we confirmed the importance of DNA binding for AHR's ability to bind to and activate Fshr promoter. Collectively, the present study demonstrates that AHR modulates Fshr transactivation by its direct association through an E-box and not by recruitment via interaction with USFs. These observations suggest that although AHR and USF may respond to different signals, they compete on binding to the same E-box. Our data, together with that from one prior study suggesting involvement of E-box motif in AHR-mediated transcription, provide novel understanding of the way in which AHR may regulate its target genes through E-box sites.


Asunto(s)
Elementos E-Box/fisiología , Regiones Promotoras Genéticas/fisiología , Receptores de Hidrocarburo de Aril/fisiología , Receptores de HFE/fisiología , Activación Transcripcional/fisiología , Animales , Secuencia de Bases , Femenino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Datos de Secuencia Molecular , Mutación/genética , Unión Proteica/fisiología , Receptores de Hidrocarburo de Aril/genética , Factores Estimuladores hacia 5'/fisiología
10.
Gen Comp Endocrinol ; 176(3): 420-5, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22240277

RESUMEN

Follicle-stimulating hormone receptor (fshr) and luteinizing hormone/choriogonadotropin receptor (lhcgr) exhibit differential temporal expression patterns during zebrafish folliculogenesis with fshr being dominant during vitellogenic growth and lhcgr increasing its expression dramatically before maturation. The dynamic and distinct expression patterns of fshr and lhcgr suggest that they are under tight regulatory control. However, the underlying mechanisms for the differential expression of the two receptors remain unknown. We have recently demonstrated that members of bone morphogenetic protein (BMP) family are largely expressed in the oocyte, while their receptors are exclusively localized on the follicle cells, suggesting a potential paracrine signaling from the oocyte to the follicle cells by BMPs. In this study, we investigated the effects of zebrafish BMP2b (zfBmp2b) and BMP4 (zfBmp4) on the expression of fshr and lhcgr using a novel co-culture approach. The recombinant zfBmp2b or zfBmp4-producing CHO cells were co-cultured with the zebrafish follicle cells followed by real-time qPCR analysis of fshr and lhcgr expression. Our results showed that zfBmp2b and zfBmp4 both down-regulated fshr, while up-regulated lhcgr expression at 24 h of co-culturing. This finding, together with the high expression level of BMP receptors in the follicle cells prior to oocyte maturation, strongly suggests a potential role for BMPs in the differential expression of fshr and lhcgr, especially in the full-grown follicles before maturation. As BMPs are largely expressed in the oocyte, this also implies an important role for the oocyte in orchestrating the differentiation and function of the follicle cells.


Asunto(s)
Proteínas Morfogenéticas Óseas/fisiología , Regulación de la Expresión Génica/fisiología , Folículo Ovárico/fisiología , Receptores de HFE/fisiología , Receptores de HL/fisiología , Pez Cebra/fisiología , Animales , Células CHO , Técnicas de Cocultivo , Cricetinae , Femenino , Oocitos/fisiología , Folículo Ovárico/citología , ARN/química , ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de HFE/genética , Receptores de HL/genética
11.
Endocrinology ; 150(1): 357-65, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18755797

RESUMEN

This report aimed to establish, using African catfish, Clarias gariepinus, as model species, a basis for understanding a well-known, although not yet clarified, feature of male fish reproductive physiology: the strong steroidogenic activity of FSHs. Assays with gonadotropin receptor-expressing cell lines showed that FSH activated its cognate receptor (FSHR) with an at least 1000-fold lower EC50 than when challenging the LH receptor (LHR), whereas LH stimulated both receptors with similar EC50s. In androgen release bioassays, FSH elicited a significant response at lower concentrations than those required to cross-activate of the LHR, indicating that FSH stimulated steroid release via FSHR-dependent mechanisms. LHR/FSHR-mediated stimulation of androgen release was completely abolished by H-89, a specific protein kinase A inhibitor, pointing to the cAMP/protein kinase A pathway as the main route for both LH- and FSH-stimulated steroid release. Localization studies showed that intratubular Sertoli cells express FSHR mRNA, whereas, as reported for the first time in a vertebrate, catfish Leydig cells express both LHR and FSHR mRNA. Testicular FSHR and LHR mRNA expression increased gradually during pubertal development. FSHR, but not LHR, transcript levels continued to rise between completion of the first wave of spermatogenesis at about 7 months and full maturity at about 12 months of age, which was associated with a previously recorded approximately 3-fold increase in the steroid production capacity per unit testis weight. Taken together, our data strongly suggest that the steroidogenic potency of FSH can be explained by its direct trophic action on FSHR-expressing Leydig cells.


Asunto(s)
Células Intersticiales del Testículo/fisiología , Receptores de HFE/fisiología , Testículo/fisiología , Andrógenos/metabolismo , Animales , Bagres/crecimiento & desarrollo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Gonadotropinas/genética , Gonadotropinas/farmacología , Masculino , Receptores de Gonadotropina/efectos de los fármacos , Receptores de Gonadotropina/fisiología , Proteínas Recombinantes/farmacología , Maduración Sexual , Testículo/crecimiento & desarrollo
12.
Reprod Biol Endocrinol ; 7: 145, 2009 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-19968884

RESUMEN

PPARgamma is highly expressed in granulosa cells by 23 days post-partum (pp) and is down-regulated in response to the LH surge. We tested the hypothesis that high levels of FSH during the neonatal period trigger the expression of PPARgamma. To determine when PPARgamma expression is initiated, ovaries were collected from neonatal rats. Messenger RNA for PPARgamma was undetectable on day 1, low from days 5-14, and increased by day 19 pp (p < 0.05). PPARgamma was detected in select granulosa cells in primary/early secondary follicles. Messenger RNA for the FSH receptor was detected as early as day 1 and remained steady throughout day 19 pp. The FSH receptor was detected by immunoblot analysis in ovaries collected 1, 2, and 5-9 days pp. In a subsequent experiment, neonatal rats were treated with acyline (GnRH antagonist) which significantly reduced FSH (p < 0.05) but not levels of mRNA for PPARgamma. The role of FSH in the induction of PPARgamma expression was further assessed in ovarian tissue from FORKO mice. Both mRNA and protein for PPARgamma were identified in ovarian tissue from FORKO mice. In summary, the FSH/FSH receptor system is present in granulosa cells prior to the onset of expression of PPARgamma. Reducing FSH during the neonatal period, or the ability to respond to FSH, did not decrease expression of mRNA for PPARgamma. These data indicate that FSH is not a primary factor initiating the expression of PPARgamma and that other agents play a role in activating its expression in the ovary.


Asunto(s)
Hormona Folículo Estimulante/fisiología , Ovario/metabolismo , PPAR gamma/biosíntesis , Animales , Animales Recién Nacidos , Western Blotting , Dieta , Femenino , Células de la Granulosa/metabolismo , Inmunohistoquímica , Ratones , Ratones Noqueados , Ensayos de Protección de Nucleasas , Embarazo , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , Radioinmunoensayo , Ratas , Ratas Sprague-Dawley , Receptores de HFE/genética , Receptores de HFE/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Gen Comp Endocrinol ; 163(3): 329-39, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19442667

RESUMEN

The gonadotropins Fsh and Lh interact with their receptors (Fshr and Lhr, respectively) in a highly specific manner in mammals with little overlap in biological activities. In fish, the biological activities seem less clearly separated considering, for example, the steroidogenic potency of both Fsh and Lh. Important determinants of the biological activity are the specificity of hormone-receptor interaction and the cellular site of receptor expression. Here, we report the pharmacological characterization of Atlantic salmon Fshr and Lhr, identify receptor-expressing cells in the ovary, and validate receptor mRNA quantification systems. For the pharmacological studies, we used highly purified coho salmon gonadotropins and found that the Fshr preferentially responded to Fsh, but was also activated by approximately 6-fold higher levels of Lh. The Lhr was specific for Lh and did not respond to Fsh. Photoperiod manipulation was used to generate ovarian tissue samples with largely differing stages of maturation. Specific real-time, quantitative (rtq) PCR assays revealed up to 40-fold (fshr) and up to 350-fold (lhr) changes in ovarian expression levels, which correlated well with the differences in ovarian weight, histology, and circulating oestrogen levels recorded in January and June, respectively. Vitellogenic ovaries were used to localise receptor-expressing cells by in situ hybridization. Granulosa cells of small and large vitellogenic follicles were positive for both receptors. Also theca cells of small and large vitellogenic follicles expressed fshr mRNA, while only in large vitellogenic follicles theca cells were (weakly) positive for lhr mRNA. While only ovulatory Lh levels seem high enough to cross-activate the Fshr, expression by both receptors by granulosa and theca cells suggests that homologous ligand receptor interaction will prevail.


Asunto(s)
Receptores de Gonadotropina/metabolismo , Salmo salar/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Línea Celular , Cromatografía Líquida de Alta Presión , Clonación Molecular , Colforsina/farmacología , Ensayo de Inmunoadsorción Enzimática , Estrógenos/sangre , Femenino , Hormona Folículo Estimulante/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hibridación in Situ , Hormona Luteinizante/farmacología , Ovario/efectos de los fármacos , Ovario/metabolismo , Receptores de HFE/genética , Receptores de HFE/metabolismo , Receptores de HFE/fisiología , Receptores de Gonadotropina/genética , Receptores de Gonadotropina/fisiología , Receptores de HL/genética , Receptores de HL/metabolismo , Receptores de HL/fisiología , Salmo salar/genética
14.
Endocrinology ; 149(7): 3279-85, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18403489

RESUMEN

Spermatogenesis in the adult male depends on the action of FSH and androgen. Ablation of either hormone has deleterious effects on Sertoli cell function and the progression of germ cells through spermatogenesis. In this study we generated mice lacking both FSH receptors (FSHRKO) and androgen receptors on the Sertoli cell (SCARKO) to examine how FSH and androgen combine to regulate Sertoli cell function and spermatogenesis. Sertoli cell number in FSHRKO-SCARKO mice was reduced by about 50% but was not significantly different from FSHRKO mice. In contrast, total germ cell number in FSHRKO-SCARKO mice was reduced to 2% of control mice (and 20% of SCARKO mice) due to a failure to progress beyond early meiosis. Measurement of Sertoli cell-specific transcript levels showed that about a third were independent of hormonal action on the Sertoli cell, whereas others were predominantly androgen dependent or showed redundant control by FSH and androgen. Results show that FSH and androgen act through redundant, additive, and synergistic regulation of spermatogenesis and Sertoli cell activity. In addition, the Sertoli cell retains a significant capacity for activity, which is independent of direct hormonal regulation.


Asunto(s)
Receptores Androgénicos/fisiología , Receptores de HFE/fisiología , Células de Sertoli/metabolismo , Espermatogénesis/fisiología , Andrógenos/farmacología , Animales , Femenino , Hormona Folículo Estimulante/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Genotipo , Masculino , Ratones , Ratones Noqueados , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Androgénicos/genética , Receptores de HFE/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células de Sertoli/efectos de los fármacos , Espermatogénesis/efectos de los fármacos , Espermatogénesis/genética
15.
Endocr Rev ; 21(5): 551-83, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11041448

RESUMEN

The recent unraveling of structures of genes for the gonadotropin subunits and gonadotropin receptors has provided reproductive endocrinologists with new tools to study normal and pathological functions of the hypothalamic-pituitary-gonadal axis. Rare inactivating mutations that produce distinctive phenotypes of isolated LH or FSH deficiency have been discovered in gonadotropin subunit genes. In addition, there is a common polymorphism in the LHbeta subunit gene with possible clinical significance as a contributing factor to pathologies of LH-dependent gonadal functions. Both activating and inactivating mutations have been detected in the gonadotropin receptor genes, a larger number in the LH receptor gene, but so far only a few in the gene for the FSH receptor. These mutations corroborate and extend our knowledge of clinical consequences of gonadotropin resistance and inappropriate gonadotropin action. The information obtained from human mutations has been complemented by animal models with disrupted or inappropriately activated gonadotropin ligand or receptor genes. These clinical and experimental genetic disease models form a powerful tool for exploring the physiology and pathophysiology of gonadotropin function and provide an excellent example of the power of molecular biological approaches in the study of pathogenesis of diseases.


Asunto(s)
Gonadotropinas Hipofisarias/genética , Mutación , Ovario/fisiología , Hipófisis/fisiología , Receptores de Gonadotropina/genética , Secuencia de Aminoácidos , Animales , Femenino , Gonadotropinas Hipofisarias/química , Gonadotropinas Hipofisarias/fisiología , Humanos , Ratones , Ratones Noqueados , Receptores de HFE/química , Receptores de HFE/genética , Receptores de HFE/fisiología , Receptores de Gonadotropina/química , Receptores de Gonadotropina/fisiología , Receptores de HL/química , Receptores de HL/genética , Receptores de HL/fisiología , Relación Estructura-Actividad
16.
Mol Hum Reprod ; 14(1): 9-15, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18084009

RESUMEN

Resistance ovarian syndrome is a heterogeneous disorder inherited as a Mendelian recessive trait and characterized by infertility, primary amenorrhea, normal karyotype and elevated serum FSH and LH levels. An inactivating mutation, C566T, in FSH receptor gene (FSHR) has been identified initially in Finland. We investigated if an adenovirus expressing a normal copy of human FSHR (Ad-hFSHR) has the ability to: (i) transfect granulosa cell lines, (ii) render the transfected cell lines responsive to FSH stimulation and (iii) transcomplement the malfunctioning form of human FSHR gene with C566T mutation. COS-7, JC-410, JC-410-P450-scc-luc and JC-410-StAR-luc cell lines were infected by Ad-hFSHR followed by treatment with FSH. Functional activity of the Ad-hFSHR was tested by measuring cyclic adenosine monophosphate (cAMP) or luciferase activity in response to FSH stimulation, and showed 2-4.6-fold increases in Ad-hFSHR transfected cells compared with untransfected or Ad-LacZ transfected cells, indicating that Ad-hFSHR is functionally active and expressing hFSHR. Generation of cAMP in cells expressing only mutated hFSHR-T566 showed minimal increase after FSH stimulation. Co-transfection of Ad-hFSHR in these cells carrying the malfunction form of human FSHR caused significant increases of 2.2-7.4-fold in FSH dependent cAMP generation (P = 0.0007). We concluded that adenovirus expressing a normal human FSHR can compensate the inactivating human FSHR-C566T mutation and restore FSH responsiveness.


Asunto(s)
Adenoviridae/genética , Terapia Genética/métodos , Mutación Puntual , Insuficiencia Ovárica Primaria/terapia , Receptores de HFE/genética , Animales , Células COS , Línea Celular , Chlorocebus aethiops , AMP Cíclico/metabolismo , Femenino , Finlandia , Hormona Folículo Estimulante/farmacología , Hormona Folículo Estimulante/uso terapéutico , Vectores Genéticos/genética , Humanos , Insuficiencia Ovárica Primaria/genética , Receptores de HFE/metabolismo , Receptores de HFE/fisiología , Transfección
17.
Mol Cell Endocrinol ; 282(1-2): 130-42, 2008 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-18248882

RESUMEN

Manipulations of mouse genome have helped to elucidate gonadotrophin function but important differences subsist between rodent and human reproduction. Studies of patients with mutations of gonadotrophins or gonadotrophin receptors genes allow understanding their physiological effects in humans. The correlation of the clinical phenotypes of patients with in vitro studies of the mutated receptor residual function and histological and immunohistological studies of the ovarian biopsies permits to understand which stages of follicular development are under FSH control. Total FSH receptor (FSHR) inactivation causes infertility with an early block of follicular maturation remarkably associated with abundant small follicles as in prepubertal ovaries and demonstrates the absolute requirement of FSH for follicular development starting from the primary stage. Partial FSHR inactivation, characterized by normal-sized ovaries, can sustain follicular development up to the early antral stages but incremental levels of FSH stimulation seem to be required for antral follicular growth before selection. These findings contrast with the traditional view of an initial gonadotrophin-independent follicular growth prior to the preantral-early antral stages. The presence of numerous reserve follicles in the ovaries of these patients may permit a future treatment of their infertility. The study of reduced FSHbeta or FSHR activity in genetically modified male mice models and in men suggests a minor impact of the FSHR on masculine fertility. Further studies on patients with a demonstrated total FSHbeta or FSHR inactivation are required to elucidate reported differences in spermatogenesis impairment. Finally, the studies of mutations of gonadotrophins and their receptors demonstrate differences in gonadotrophin function between genetically modified rodents and humans which suggest prudence in extrapolating observations in rodents to human reproduction. Ovarian hyperstimulation syndrome (OHSS) can infrequently arise spontaneously during pregnancy, but most often it is an iatrogenic complication of ovarian stimulation treatments with ovulation drugs for in vitro fertilization. The first genetic cause of familial recurrent spontaneous OHSS was identified as a broadening specificity of the FSHR for hCG due to naturally occurring heterozygous mutations located unexpectedly in the transmembrane domain of the FSHR. Broadening specificity of a G protein-coupled receptor is extremely rare. These observations led to the identification of the etiology of this previously unexplained syndrome and permitted to conceive novel models of FSHR activation. Susceptibility to iatrogenic OHSS or its clinical severity may be associated with FSHR polymorphisms with slightly different activities in vivo as suggested by several studies. The study of larger cohorts is needed to evaluate the clinical impact of these observations in the management of patients undergoing IVF protocols.


Asunto(s)
Mutación/genética , Receptores de HFE/genética , Receptores de HFE/fisiología , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Infertilidad Femenina/genética , Infertilidad Masculina/genética , Masculino , Ratones , Síndrome de Hiperestimulación Ovárica/genética , Linaje
18.
Mol Endocrinol ; 21(2): 574-80, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17110401

RESUMEN

Comparison between wild-type and mutated glycoprotein hormone receptors (GPHRs), TSH receptor, FSH receptor, and LH-chorionic gonadotropin receptor is established to identify determinants involved in molecular activation mechanism. The basic aims of the current work are 1) the discrimination of receptor phenotypes according to the differences between activity states they represent, 2) the assignment of classified phenotypes to three-dimensional structural positions to reveal 3) functional-structural hot spots and 4) interrelations between determinants that are responsible for corresponding activity states. Because it is hard to survey the vast amount of pathogenic and site-directed mutations at GPHRs and to improve an almost isolated consideration of individual point mutations, we present a system for systematic and diversified sequence-structure-function analysis (http://www.fmp-berlin.de/ssfa). To combine all mutagenesis data into one set, we converted the functional data into unified scaled values. This at least enables their comparison in a rough classification manner. In this study we describe the compiled data set and a wide spectrum of functions for user-driven searches and classification of receptor functionalities such as cell surface expression, maximum of hormone binding capability, and basal as well as hormone-induced Galphas/Galphaq mediated cAMP/inositol phosphate accumulation. Complementary to known databases, our data set and bioinformatics tools allow functional and biochemical specificities to be linked with spatial features to reveal concealed structure-function relationships by a semiquantitative analysis. A comprehensive discrimination of specificities of pathogenic mutations and in vitro mutant phenotypes and their relation to signaling mechanisms of GPHRs demonstrates the utility of sequence-structure-function analysis. Moreover, new interrelations of determinants important for selective G protein-mediated activation of GPHRs are resumed.


Asunto(s)
Bases de Datos de Proteínas , Receptores de HFE/química , Receptores de HL/química , Receptores de Tirotropina/química , Animales , Humanos , Modelos Moleculares , Mutación , Conformación Proteica , Receptores de HFE/genética , Receptores de HFE/fisiología , Receptores de HL/genética , Receptores de HL/fisiología , Receptores de Tirotropina/genética , Receptores de Tirotropina/fisiología , Transducción de Señal , Relación Estructura-Actividad
19.
Theriogenology ; 69(1): 72-80, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17980420

RESUMEN

The study of follicular dynamics began in the mid-20th century, but progress has been particularly rapid in the last two decades through the use of tools that have enabled serial, non-invasive examination. A brief overview of early oogenesis and folliculogenesis is provided as a backdrop to the evolution of our understanding of follicular dynamics during the bovine estrous cycle. Studies to date support the concept that the pair of ovaries acts as a single unit and influences follicular development primarily via systemic endocrine routes involving ovarian and uterine products, the gonadotropins, and their receptors. Dominant and subordinate follicles pass through growing, static and regressing phases that have distinct morphologic and biochemical characteristics; these changes are the basis of efforts focused on diagnosing and manipulating follicular status. An update of research progress highlights recent findings on the repeatability (predictability) within individuals of follicle recruitment and wave pattern (two- versus three-wave cycles), the relationship between oocyte competence and follicular status, and the dynamics of small follicles. Recent studies documented that wave emergence and follicular dominance are apparent earlier than previously reported, and on the basis of periodic endogenous FSH surges and the presence of FSH receptors, the hypothesis that follicles become progressively entrained to waves from the earliest stages of development is introduced. Lastly, recent studies comparing old cows and their young daughters provide a new understanding of the effects of aging on gonadotropins and ovarian steroids, follicular dynamics, ovarian response to synchronization, superstimulation, and oocyte competence.


Asunto(s)
Bovinos/fisiología , Folículo Ovárico/fisiología , Animales , Ciclo Estral/fisiología , Femenino , Hormona Folículo Estimulante/fisiología , Oocitos/fisiología , Folículo Ovárico/diagnóstico por imagen , Receptores de HFE/fisiología , Ultrasonografía
20.
Bull Mem Acad R Med Belg ; 163(10-12): 481-4; discussion 484-5, 2008.
Artículo en Francés | MEDLINE | ID: mdl-20120254

RESUMEN

The constitutive activation of the follitropin receptor (FSHR) could lead to promiscuous toxic activation by placental chorionic gonadotropin (CG) during human pregnancy. This study demonstrated that the evolution of the transmembrane region of the simian FSHR parallels the progressive accumulation of CG copies in the primate genomes. We assist to a purifying selection to keep the FSHR constitutively inactive and thus insensitive to CG.


Asunto(s)
Gonadotropina Coriónica/fisiología , Evolución Molecular , Selección Genética , Animales , Gonadotropina Coriónica/genética , Humanos , Primates , Receptores de HFE/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA