Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Biol Sex Differ ; 8(1): 24, 2017 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-28693572

RESUMO

BACKGROUND: The thermoneutral zone (TNZ) is a species-specific range of ambient temperature (T a), at which mammals can maintain a constant body temperature with the lowest metabolic rate. The TNZ for an adult mouse is between 26 and 34 °C. Interestingly, female mice prefer a higher T a than male mice although the underlying mechanism for this sex difference is unknown. Here, we tested whether gonadal hormones are dominant factors controlling temperature preference in male and female mice. METHODS: We performed a temperature preference test in which 10-week-old gonadectomized and sham-operated male and female C57BL/6J mice were allowed to choose to reside at the thermoneutral cage of 29 °C or an experimental cage of 26, 29, or 32 °C. RESULTS: All mice preferred a T a higher than 26 °C, especially in the inactive phase. Choosing between 29 and 32 °C, female mice resided more at 32 °C while male mice had no preference between the temperatures. Hence, the preferred T a for female mice was significantly higher (0.9 ± 0.2 °C) than that for male mice. However, gonadectomy did not influence the T a preference. CONCLUSIONS: Female mice prefer a warmer environment than male mice, a difference not affected by gonadectomy. This suggests that thermal-sensing mechanisms may be influenced by sex-specific pathways other than gonadal factors or that the thermoregulatory set point has already been determined prior to puberty.


Assuntos
Hormônios/metabolismo , Caracteres Sexuais , Temperatura , Animais , Regulação da Temperatura Corporal/fisiologia , Peso Corporal/fisiologia , Comportamento Alimentar/fisiologia , Feminino , Masculino , Camundongos Endogâmicos C57BL , Atividade Motora/fisiologia , Orquiectomia , Ovariectomia , Distribuição Aleatória , Comportamento Espacial/fisiologia , Sensação Térmica/fisiologia
2.
Fertil Steril ; 103(4): 1081-1088.e3, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25721191

RESUMO

OBJECTIVE: To assess whether an FSH receptor polymorphism (Asn680Ser, rs6166) can affect the outcome of ovulation induction in normogonadotropic (World Health Organization class 2 [WHO2]) anovulatory subfertile women. DESIGN: Prospective, longitudinal, cohort study. SETTING: University-based fertility unit. PATIENT(S): A total of 240 consecutive women diagnosed with WHO2 anovulatory subfertility who underwent ovulation induction therapy. Results were replicated in a retrospective cohort of 185 patients with polycystic ovary syndrome (PCOS) (Rotterdam criteria). INTERVENTION(S): Ovulation induction using clomiphene citrate (CC) as first-line and exogenous gonadotropins (exFSH) as second-line therapy. MAIN OUTCOME MEASURE(S): Clomiphene-resistant anovulation (CRA), clomiphene failure (CCF), and ongoing pregnancy rate. RESULT(S): Genotyped patients (n = 159) were similar to nongenotyped women (n = 81) regarding clinical characteristics and outcomes of ovulation induction. The 680(Ser) allele was associated with CRA. A pooled analysis of both cohorts showed an 89% higher chance of CRA after CC treatment (odds ratio 1.9 [95% confidence interval 1.1-3.3]) in homozygous carriers of the FSH receptor variant (680(Ser/Ser)). A lower chance of ongoing pregnancy (hazard ratio 0.51 [95% confidence interval 0.27-0.98]) was observed among these patients during CC treatment in the prospective cohort. CONCLUSION(S): An FSH receptor polymorphism is associated with CRA during treatment with clomiphene citrate. These data may be used to design a treatment algorithm that is more efficacious and better tailored to the individual patient.


Assuntos
Anovulação/genética , Anovulação/terapia , Infertilidade Feminina/genética , Infertilidade Feminina/terapia , Indução da Ovulação , Polimorfismo de Nucleotídeo Único , Receptores do FSH/genética , Adulto , Anovulação/classificação , Clomifeno/uso terapêutico , Resistência a Medicamentos/genética , Feminino , Fármacos para a Fertilidade Feminina/uso terapêutico , Humanos , Infertilidade Feminina/classificação , Síndrome do Ovário Policístico/genética , Síndrome do Ovário Policístico/terapia , Gravidez , Estudos Retrospectivos , Resultado do Tratamento , Organização Mundial da Saúde , Adulto Jovem
3.
Mol Biol Rep ; 41(12): 7967-72, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25163632

RESUMO

The melanocortin 4 receptor (MC4R) is expressed in the hypothalamus and is essential for regulation of appetite and energy expenditure. MC4R dysfunction in humans causes hyperphagia, impaired satiety and obesity. We have identified a novel c.216C>A (N72 K) homozygous mutation in MC4R in a girl with severe obesity. The patient presented with early-onset obesity and hyperphagia indicating an effect of the homozygous mutation on her phenotype. In silico analyses indicate a damaging effect on receptor function, and the mutation is unusual in occurring in the first intra-cellular loop of the receptor. Site-directed mutagenesis was used to generate plasmid constructs expressing wild-type and mutant MC4R. These were transfected into HEK293 cells and assessed for cAMP responsiveness to α-MSH. Cells expressing N-terminal HA and C-terminal GFP-tagged MC4R were assessed by immunofluorescence confocal microscopy and flow cytometry for correct cell-surface localization. The maximal response of the mutant MC4R to α-MSH was decreased to 20 ± 1 % of the wild type receptor response, and the EC50 was increased from 16.5 ± 5.4 nM to 37.0 ± 8.3 nM. Localization of N- and C-terminally tagged MC4R by confocal microscopy and flow cytometry showed aberrant retention of the mutant receptor in the cytoplasm. Our data describe a rare homozygous inactivating mutation in the first intra-cellular loop of MC4R that markedly impairs its function and is associated with early-onset obesity and hyperphagia.


Assuntos
Homozigoto , Mutação de Sentido Incorreto , Obesidade Infantil/genética , Receptor Tipo 4 de Melanocortina/genética , Feminino , Células HEK293 , Humanos , Hiperfagia/genética , Lactente , alfa-MSH/farmacologia
4.
Mol Cell Endocrinol ; 394(1-2): 99-104, 2014 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-25017734

RESUMO

Cushing's disease, a hypercortisolemic state induced by an ACTH overexpressing pituitary adenoma, causes increased morbidity and mortality. Selective antagonism of the melanocortin type 2 receptor (MC2R) may be a novel treatment modality. Five structurally related peptides with modified HFRW sites but intact putative MC2R binding sites were tested for antagonistic activity at MC1R, MC2R/MRAP, MC3R, MC4R and MC5R. Two of these peptides (GPS1573 and GPS1574) dose-dependently antagonized ACTH-stimulated MC2R activity (IC50s of 66±23 nM and 260±1 nM, respectively). GPS1573 and 1574 suppressed the Rmax but not EC50 of ACTH on MC2R, indicating non-competitive antagonism. These peptides did not antagonize α-MSH stimulation of MC1R and antagonized MC3, 4 and 5R at markedly lower potency. GP1573 and GPS1574 antagonize MC4R with IC50s of 950 nM and 3.7 µM, respectively. In conclusion, two peptide antagonists were developed with selectivity for MC2R, forming a platform for development of a medical treatment for Cushing's disease.


Assuntos
Hormônio Adrenocorticotrópico/farmacologia , Desenho de Fármacos , Peptídeos/síntese química , Receptor Tipo 2 de Melanocortina/antagonistas & inibidores , Hormônio Adrenocorticotrópico/genética , Hormônio Adrenocorticotrópico/metabolismo , Sequência de Aminoácidos , Relação Dose-Resposta a Droga , Expressão Gênica , Células HEK293 , Humanos , Dados de Sequência Molecular , Peptídeos/farmacologia , Hipersecreção Hipofisária de ACTH/tratamento farmacológico , Ligação Proteica , Receptor Tipo 1 de Melanocortina/química , Receptor Tipo 1 de Melanocortina/genética , Receptor Tipo 1 de Melanocortina/metabolismo , Receptor Tipo 2 de Melanocortina/química , Receptor Tipo 2 de Melanocortina/genética , Receptor Tipo 2 de Melanocortina/metabolismo , Receptor Tipo 3 de Melanocortina/química , Receptor Tipo 3 de Melanocortina/genética , Receptor Tipo 3 de Melanocortina/metabolismo , Receptor Tipo 4 de Melanocortina/química , Receptor Tipo 4 de Melanocortina/genética , Receptor Tipo 4 de Melanocortina/metabolismo , Receptores de Melanocortina/química , Receptores de Melanocortina/genética , Receptores de Melanocortina/metabolismo , Relação Estrutura-Atividade , Transfecção
5.
Endocrinology ; 153(6): 2861-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22334715

RESUMO

Polycystic ovary syndrome (PCOS), the most common endocrine disorder in women in their reproductive age, is characterized by both reproductive and metabolic features. Recent studies in human, nonhuman primates, and sheep suggest that hyperandrogenism plays an important role in the development of PCOS. We investigated whether chronic dihydrotestosterone (DHT) exposure in mice reproduces both features of PCOS. Such a model would allow us to study the mechanism of association between the reproductive and metabolic features in transgenic mice. In this study, prepubertal female mice received a 90 d continuous release pellet containing the nonaromatizable androgen DHT or vehicle. At the end of the treatment period, DHT-treated mice were in continuous anestrous, their ovaries contained an increased number of atretic follicles, with the majority of atretic antral follicles having a cyst-like structure. Chronic DHT-exposed mice had significantly higher body weights (21%) than vehicle-treated mice. In addition, fat depots of DHT-treated mice displayed an increased number of enlarged adipocytes (P < 0.003). Leptin levels were elevated (P < 0.013), adiponectin levels were diminished (P < 0.001), and DHT-treated mice were glucose intolerant (P < 0.001). In conclusion, a mouse model of PCOS has been developed showing reproductive and metabolic characteristics associated with PCOS in women.


Assuntos
Modelos Animais de Doenças , Metabolismo dos Lipídeos/fisiologia , Síndrome do Ovário Policístico/fisiopatologia , Reprodução/fisiologia , Adiponectina/sangue , Animais , Peso Corporal/fisiologia , Di-Hidrotestosterona , Ensaio de Imunoadsorção Enzimática , Feminino , Expressão Gênica , Humanos , Imuno-Histoquímica , Insulina/sangue , Leptina/sangue , Hormônio Luteinizante/sangue , Camundongos , Camundongos Endogâmicos C57BL , Folículo Ovariano/metabolismo , Folículo Ovariano/patologia , Ovário/metabolismo , Ovário/patologia , Fenótipo , Síndrome do Ovário Policístico/induzido quimicamente , Síndrome do Ovário Policístico/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
Int J Endocrinol ; 2012: 237084, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22253624

RESUMO

Disorder of sex development (DSD) patients in Indonesia most often do not receive a proper diagnostic evaluation and treatment. This study intended to categorize 88 Indonesian patients in accordance with the new consensus DSD algorithm. Diagnostic evaluation including clinical, hormonal, genetic, imaging, surgical, and histological parameters was performed. Fifty-three patients were raised as males, and 34 as females. Of 22 patients with 46, XX DSD, 15 had congenital adrenal hyperplasia, while in one patient, an ovarian Leydig cell tumor was found. In all 58 46, XY DSD patients, 29 were suspected of a disorder of androgen action (12 with an androgen receptor mutation), and in 9, gonadal dysgenesis was found and, in 20, severe hypospadias e.c.i. Implementation of the current consensus statement in a resource-poor environment is very difficult. The aim of the diagnostic workup in developing countries should be to end up with an evidence-based diagnosis. This is essential to improve treatment and thereby to improve the patients' quality of life.

7.
Nat Rev Endocrinol ; 8(6): 331-41, 2012 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-22231848

RESUMO

Primary ovarian insufficiency (POI), also known as premature ovarian failure, is a disorder of infertility characterized by amenorrhoea, low estrogen levels and increased gonadotropin levels in women aged <40 years. POI is the result of premature exhaustion of the follicle pool or can be attributed to follicular dysfunction, for example, owing to mutations in the follicle-stimulating hormone receptor or steroidogenic cell autoimmunity. Moreover, advances in cancer therapeutics over the past decades have led to increasing survival rates for both paediatric and adult malignancies. Given the gonadotoxic effect of many cancer treatments, more women develop POI. A marker that predicts whether women are at risk of POI would, therefore, aid in early diagnosis and fertility counselling. Anti-Müllerian hormone (AMH), a growth factor produced solely by small, growing follicles in the ovary, might constitute such a marker, as serum levels of this hormone correlate strongly with the number of growing follicles. In addition, AMH could potentially help assess the progression of ovarian senescence, as serum AMH levels are independent of hypothalamic-pituitary-gonadal axis function and decrease to undetectable levels at menopause. In cancer survivors, serum AMH levels correlate with the extent of gonadal damage. In this Review, we provide an overview of the current studies that have measured AMH in women with POI of various aetiologies and discuss its possible application as a marker to determine ovarian reserve.


Assuntos
Hormônio Antimülleriano/sangue , Ovário/fisiopatologia , Insuficiência Ovariana Primária/sangue , Hormônio Antimülleriano/fisiologia , Biomarcadores/sangue , Feminino , Humanos , Folículo Ovariano/fisiopatologia , Insuficiência Ovariana Primária/etiologia , Insuficiência Ovariana Primária/fisiopatologia
8.
Fertil Steril ; 96(2): 459-63, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21719005

RESUMO

OBJECTIVE: To describe changes of anti-Müllerian hormone (AMH) and inhibin B during low-dose gonadotropin ovulation induction (OI) treatment in women with polycystic ovary syndrome (PCOS), and thus disturbed selection of the dominant follicle. DESIGN: Observational study. SETTING: A referral fertility clinic. PATIENT(S): Women with PCOS (n = 48) and normo-ovulatory women (n = 23). INTERVENTION(S) AND MAIN OUTCOME MEASURE(S): Serum AMH, inhibin B, FSH, and E(2) concentrations were measured at start of stimulation, on the day of follicle selection, and at administration of hCG during OI cycles and were compared with concentration measured during the normal menstrual cycle. RESULT(S): Development of a single dominant follicle was observed in 92% of all OI cycles, reflected by similar E(2) concentrations compared with those in spontaneous cycles. AMH concentrations were constant during low-dose ovarian stimulation. Inhibin B concentrations remained elevated in patients with PCOS, suggesting prolonged survival of small antral follicles, whereas in controls inhibin B concentrations declined during the late follicular phase. CONCLUSION(S): The lack of change in AMH and inhibin B concentrations suggest that follicle dynamics during low-dose stimulation seem different from those during controlled ovarian hyperstimulation. In addition, constant AMH and inhibin B levels suggest that neither AMH nor inhibin B is an accurate marker of ovarian response after low-dose gonadotropin OI in patients with PCOS.


Assuntos
Hormônio Antimülleriano/sangue , Fármacos para a Fertilidade Feminina/uso terapêutico , Hormônio Foliculoestimulante Humano/uso terapêutico , Inibinas/sangue , Indução da Ovulação , Ovulação/efeitos dos fármacos , Síndrome do Ovário Policístico/tratamento farmacológico , Adulto , Análise de Variância , Biomarcadores/sangue , Estudos de Casos e Controles , Estradiol/sangue , Feminino , Fármacos para a Fertilidade Feminina/administração & dosagem , Fármacos para a Fertilidade Feminina/sangue , Hormônio Foliculoestimulante Humano/administração & dosagem , Hormônio Foliculoestimulante Humano/sangue , Humanos , Países Baixos , Síndrome do Ovário Policístico/sangue , Síndrome do Ovário Policístico/fisiopatologia , Proteínas Recombinantes/uso terapêutico , Fatores de Tempo , Resultado do Tratamento , Adulto Jovem
9.
J Clin Endocrinol Metab ; 96(7): E1197-205, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21490077

RESUMO

CONTEXT: Germline and somatic activating mutations in the LH receptor (LHR) gene have been reported. OBJECTIVE: Our objective was to perform mutation analysis of the LHR gene of patients with Leydig cell adenoma or hyperplasia. Functional studies were conducted to compare the D578H-LHR mutant with the wild-type (WT)-LHR and the D578G-LHR mutant, a classic cause of testotoxicosis. The three main signal transduction pathways in which LHR is involved were studied. PATIENTS: We describe eight male patients with gonadotropin-independent precocious puberty due to Leydig cell adenoma or hyperplasia. RESULTS: The D578H-LHR mutation was found in the adenoma or nodule with hyperplasia in all but two patients. D578H-LHR displayed a constitutively increased but noninducible production of cAMP, led to a very high production of inositol phosphates, and induced a slight phosphorylation of p44/42 MAPK in the absence of human chorionic gonadotropin. The D578G-LHR showed a response intermediate between WT-LHR and the D578H-LHR. Subcellular localization studies showed that the WT-LHR was almost exclusively located at the cell membrane, whereas the D578H-LHR showed signs of internalization. D578H-LHR was the only receptor to colocalize with early endosomes in the absence of human chorionic gonadotropin. CONCLUSIONS: Although several LHR mutations have been reported in testotoxicosis, the D578H-LHR mutation, which has been found only as a somatic mutation, appears up until now to be specifically responsible for Leydig cell adenomas. This is reflected by the different activation of the signal transduction pathways, when compared with the WT-LHR or D578G-LHR, which may explain the tumorigenesis in the D578H mutant.


Assuntos
Adenoma/genética , Hiperplasia/genética , Células Intersticiais do Testículo/patologia , Puberdade Precoce/genética , Receptores do LH/genética , Neoplasias Testiculares/genética , Adenoma/metabolismo , Adenoma/patologia , Criança , Pré-Escolar , Análise Mutacional de DNA , Humanos , Hiperplasia/metabolismo , Hiperplasia/patologia , Células Intersticiais do Testículo/metabolismo , Masculino , Mutação , Puberdade Precoce/metabolismo , Puberdade Precoce/patologia , Receptores do LH/metabolismo , Neoplasias Testiculares/metabolismo , Neoplasias Testiculares/patologia
10.
Mol Cell Endocrinol ; 302(1): 58-64, 2009 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-19356624

RESUMO

Worldwide, breast cancer is the most frequently occurring malignancy in women. Early age at full term pregnancy has a protective effect against breast cancer. Evidence coming from a rat breast cancer model suggests a possible role for the pregnancy hormone hCG, a ligand of the LH receptor, as a mediator for this effect. In a previous study, we found that a common polymorphism in the LH receptor associates with tumor progression in premenopausal breast cancer patients, as carriers of the variant receptor showed a shorter disease free survival compared to non-carriers. How hCG and its receptor exert their effects on breast cancer, however, is unclear. One possibility is that these effects take place through LH receptors present in the ovaries, thereby influencing steroid hormone production. Another possibility is that the effects take place through LH receptors present in breast tumor cells themselves, as some studies have detected the receptor in both normal and neoplastic breast tissues and in breast cancer cell lines. To investigate whether a direct effect of LH signaling in breast cancer is likely, we measured LH receptor mRNA expression levels in 1551 breast tumors and 42 different human breast cancer cell lines using a qRT-PCR with a wide dynamic range. In addition, associations between LH receptor expression and clinico-pathologic factors were investigated. Assay validation showed that as little as ?10 copies per reaction volume of LH receptor cDNA could still be detected by our assay. We show that LH receptors are undetectable in 62% of breast tumor samples and 41 of 42 breast cancer cell lines. For the remaining samples we found expression levels to be very low. Although low, expression of the LH receptor appears to be associated with normal breast cells, favorable tumor characteristics and low tumor percentage. Since expression of the LH receptor in breast cancer cells is very low, it almost excludes the possibility of direct signaling effects. We therefore conclude that signaling effects of the LH receptor on breast cancer most likely take place by an indirect pathway through the ovaries.


Assuntos
Neoplasias da Mama/metabolismo , Regulação Neoplásica da Expressão Gênica , Receptores do LH/metabolismo , Adulto , Idoso , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/metabolismo , Receptores do LH/genética , Reprodutibilidade dos Testes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
11.
Hum Reprod ; 24(1): 241-9, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18854405

RESUMO

BACKGROUND: Polycystic ovaries display an increased number of pre-antral and antral follicles compared with normal ovaries, suggesting that early and late follicle development are disturbed. The pathophysiology of this process is poorly understood. Since the transforming growth factor beta family members, anti-Müllerian hormone (AMH) and bone morphogenetic proteins (BMPs), inhibit FSH sensitivity, their signalling may contribute to the aberrant follicle development in these women. Here, we investigated the role of ALK2, a type I receptor for AMH/BMP signalling, in PCOS using a genetic approach. METHODS: Seven single nucleotide polymorphisms in the ACVR1 gene, encoding ALK2, were genotyped in 359 PCOS patients and 30 normo-ovulatory and 3543 population-based control women, and haplotypes were determined. Subsequently, the association of ACVR1 variants with ovarian parameters and hormone levels was investigated. RESULTS: The polymorphisms rs1220134, rs10497189 and rs2033962 and their corresponding haplotypes did not show different frequencies from controls, but were associated with AMH levels in PCOS women (P = 0.001, P = 0.002 and P = 0.007, respectively). Adjustment for follicle number revealed that the association with AMH levels was, in part, independent from follicle number, suggesting that variants in ACVR1 also influence AMH production per follicle. CONCLUSIONS: Genetic variation within ACVR1 is associated with AMH levels and follicle number in PCOS women, suggesting that ALK2 signalling contributes to the disturbed folliculogenesis in PCOS patients.


Assuntos
Receptores de Ativinas Tipo I/genética , Hormônio Antimülleriano/metabolismo , Síndrome do Ovário Policístico/genética , Polimorfismo de Nucleotídeo Único , Receptores de Ativinas Tipo I/fisiologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Hormônio Antimülleriano/genética , Estudos de Coortes , Feminino , Frequência do Gene , Haplótipos , Humanos , Desequilíbrio de Ligação , Pessoa de Meia-Idade , Folículo Ovariano/metabolismo , Folículo Ovariano/fisiologia , Fatores de Risco , Transdução de Sinais
12.
Trends Endocrinol Metab ; 19(9): 340-7, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18805020

RESUMO

Anti-Müllerian hormone (AMH) has important roles in postnatal ovarian function. Produced by ovarian granulosa cells, AMH is involved in initial follicle development. In fact, serum AMH level correlates with ovarian follicle number. In patients with polycystic ovary syndrome (PCOS), AMH levels are elevated, which indicates its potential relevance in PCOS diagnosis and management. AMH represents a useful clinical marker for the assessment of ovarian reserve in cases of subfertility caused by advanced age in women. A potential role for AMH in dominant follicle selection has also been suggested. Future challenges comprise the availability of a well-standardized assay and the development of AMH agonists and antagonists as possible tools to manipulate ovarian function for contraception or ovarian longevity.


Assuntos
Hormônio Antimülleriano/metabolismo , Ovário/fisiopatologia , Hormônio Antimülleriano/agonistas , Hormônio Antimülleriano/antagonistas & inibidores , Feminino , Humanos , Modelos Biológicos , Folículo Ovariano/efeitos dos fármacos , Folículo Ovariano/metabolismo , Folículo Ovariano/fisiopatologia , Ovário/efeitos dos fármacos , Ovário/metabolismo , Síndrome do Ovário Policístico/tratamento farmacológico , Síndrome do Ovário Policístico/metabolismo , Síndrome do Ovário Policístico/fisiopatologia
13.
J Biol Chem ; 283(38): 25821-8, 2008 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-18641392

RESUMO

The luteinizing hormone (LH) receptor plays an essential role in male and female gonadal function. Together with the follicle-stimulating hormone (FSH) and thyroid stimulating hormone (TSH) receptors, the LH receptor forms the family of glycoprotein hormone receptors. All glycoprotein hormone receptors share a common modular topography, with an N-terminal extracellular ligand binding domain and a C-terminal seven-transmembrane transduction domain. The ligand binding domain consists of 9 leucine-rich repeats, flanked by N- and C-terminal cysteine-rich regions. Recently, crystal structures have been published of the extracellular domains of the FSH and TSH receptors. However, the C-terminal cysteine-rich region (CCR), also referred to as the "hinge region," was not included in these structures. Both structure and function of the CCR therefore remain unknown. In this study we set out to characterize important domains within the CCR of the LH receptor. First, we mutated all cysteines and combinations of cysteines in the CCR to identify the most probable disulfide bridges. Second, we exchanged large parts of the LH receptor CCR by its FSH receptor counterparts, and characterized the mutant receptors in transiently transfected HEK 293 cells. We zoomed in on important regions by focused exchange and deletion mutagenesis followed by alanine scanning. Mutations in the CCR specifically decreased the potencies of LH and hCG, because the potency of the low molecular weight agonist Org 41841 was unaffected. Using this unbiased approach, we identified Asp(330) and Tyr(331) as key amino acids in LH/hCG mediated signaling.


Assuntos
Ácido Aspártico/química , Receptores do LH/química , Receptores do LH/metabolismo , Tirosina/química , Sequência de Aminoácidos , Linhagem Celular , Cisteína/química , Dissulfetos , Genes Reporter , Humanos , Modelos Biológicos , Dados de Sequência Molecular , Mutação , Conformação Proteica , Estrutura Terciária de Proteína , Receptores do FSH/metabolismo
14.
J Clin Endocrinol Metab ; 93(4): 1310-6, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18230658

RESUMO

CONTEXT: The common characteristic of polycystic ovary syndrome (PCOS) is a disturbance in the selection of the dominant follicle, resulting in anovulation. In PCOS women, serum anti-Müllerian hormone (AMH) levels are elevated. Because AMH decreases FSH sensitivity in mice, the elevated AMH levels may contribute to the disturbed follicle selection in PCOS women. OBJECTIVE: The objective of the study was to investigate the role of the AMH signaling pathway in the pathophysiology of PCOS using a genetic approach. DESIGN: The association of the AMH Ile(49)Ser (rs10407022) and the AMH type II receptor -482 A>G (rs2002555) polymorphism with PCOS susceptibility and phenotype was studied in a large cohort of PCOS women. SETTING/SUBJECTS: A total of 331 women with PCOS, 32 normoovulatory controls, and 3635 population-based controls were included. MAIN OUTCOME MEASURES: Ovarian parameters, serum AMH, FSH, androgen, and estradiol levels were measured. RESULTS: Genotype and allele frequencies for the AMH Ile(49)Ser and AMH type II receptor -482 A>G polymorphism were similar in PCOS women and controls. However, within the group of PCOS women, carriers of the AMH (49)Ser allele less often had polycystic ovaries (92.7 vs. 99.5%, P = 0.0004), lower follicle numbers (P = 0.03), and lower androgen levels, compared with noncarriers (P = 0.04). In addition, in vitro studies demonstrated that the bioactivity of the AMH (49)Ser protein is diminished, compared with the AMH (49)Ile protein (P < 0.0001). CONCLUSIONS: Genetic variants in the AMH and AMH type II receptor gene do not influence PCOS susceptibility. However, our results suggest that the AMH Ile(49)Ser polymorphism contributes to the severity of the PCOS phenotype.


Assuntos
Androgênios/sangue , Hormônio Antimülleriano/genética , Folículo Ovariano/patologia , Síndrome do Ovário Policístico/genética , Polimorfismo Genético , Adulto , Animais , Hormônio Antimülleriano/sangue , Linhagem Celular , Feminino , Genótipo , Humanos , Camundongos , Síndrome do Ovário Policístico/sangue , Síndrome do Ovário Policístico/patologia , Receptores de Peptídeos/genética , Receptores de Fatores de Crescimento Transformadores beta/genética
15.
J Clin Endocrinol Metab ; 92(10): 3869-74, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17726078

RESUMO

PURPOSE: The aim of this study was to evaluate the long-term effects of combination chemotherapy treatment for girls with Hodgkin's lymphoma (HL) on gonadal function using anti-Müllerian hormone (AMH) and inhibin B as ovarian reserve parameters. PATIENTS AND METHODS: LH, FSH, inhibin B, and AMH were measured in 32 women treated from 1974 to 1998 for pediatric HL with chemotherapy, with the intention to avoid radiotherapy. All patients [median age 25.0 yr (range 19.2-40.4 yr)] were in complete remission with a median follow-up time of 14.0 yr (range 5.7-24.5 yr) after therapy. All patients were treated with combination chemotherapy doxorubicin, bleomycin, vinblastine and dacarbazine (ABVD) or EBVD with or without mechlorethamine, vincristine, procarbazine, and prednisone (MOPP). Because of incomplete remission or relapse, involved field radiotherapy was needed in seven of 32 women. Results were compared with a healthy control group. RESULTS: Patients treated with six or more cycles of MOPP combination chemotherapy had significantly higher levels of FSH and lower serum levels of inhibin B and AMH, compared with healthy women [FSH, 17.0 vs. 6.0 U/liter (P < 0.05); inhibin B, 23.0 vs. 112.5 ng/liter (P < 0.01); AMH, 0.39 vs. 2.10 microg/liter (P < 0.01)]. AMH was also significantly lower, compared with women treated without MOPP (median 0.39 vs. 1.40 microg/liter; P = 0.01). CONCLUSIONS: Women treated during childhood for HL with MOPP seem to have a distinctly lower ovarian reserve as measured by lower AMH values at early adulthood, compared with healthy women. Moreover, AMH seems to be the only predictor that is sufficiently sensitive to detect this decrease in ovarian reserve.


Assuntos
Hormônio Antimülleriano/sangue , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Biomarcadores/sangue , Doença de Hodgkin/tratamento farmacológico , Insuficiência Ovariana Primária/induzido quimicamente , Adolescente , Adulto , Fatores Etários , Idade de Início , Bleomicina/efeitos adversos , Criança , Dacarbazina/efeitos adversos , Doxorrubicina/efeitos adversos , Epirubicina/efeitos adversos , Feminino , Seguimentos , Humanos , Masculino , Mecloretamina/efeitos adversos , Ovário/efeitos dos fármacos , Ovário/fisiologia , Valor Preditivo dos Testes , Prednisona/efeitos adversos , Gravidez , Insuficiência Ovariana Primária/sangue , Insuficiência Ovariana Primária/diagnóstico , Procarbazina/efeitos adversos , Indução de Remissão , Vimblastina/efeitos adversos , Vincristina/efeitos adversos
16.
Breast Cancer Res ; 9(4): R51, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17692113

RESUMO

BACKGROUND: Breast cancer development and progression are dependent on estrogen activity. In premenopausal women, estrogen production is mainly regulated through the hypothalamic-pituitary-gonadal (HPG) axis. METHODS: We have investigated the prognostic significance of two variants of genes involved in the HPG-axis, the GnRH (encoding gonadotropin-releasing hormone) 16Trp/Ser genotype and the LHR (encoding the luteinizing hormone receptor) insLQ variant, in retrospectively collected premenopausal breast cancer patients with a long follow-up (median follow-up of 11 years for living patients). RESULTS: Carriership was not related with breast cancer risk (the case control study encompassed 278 premenopausal cases and 1,758 premenopausal controls). A significant adverse relationship of the LHR insLQ and GnRH 16Ser genotype with disease free survival (DFS) was observed in premenopausal (hormone receptor positive) breast cancer patients. In particular, those patients carrying both the GnRH 16Ser and LHR insLQ allele (approximately 25%) showed a significant increased risk of relapse, which was independent of traditional prognostic factors (hazard ratio 2.14; 95% confidence interval 1.32 to 3.45; P = 0.002). CONCLUSION: We conclude that the LHR insLQ and GnRH 16Ser alleles are independently associated with shorter DFS in premenopausal patients. When validated, these findings may provide a lead in the development of tailored treatment for breast cancer patients carrying both polymorphisms.


Assuntos
Neoplasias da Mama/genética , Hormônio Liberador de Gonadotropina/genética , Polimorfismo Genético , Receptores do LH/genética , Adulto , Antineoplásicos Hormonais/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/cirurgia , Estudos de Casos e Controles , Intervalo Livre de Doença , Feminino , Genótipo , Humanos , Mastectomia Segmentar , Pessoa de Meia-Idade , Pré-Menopausa , Receptores de Estrogênio , Estudos Retrospectivos , Taxa de Sobrevida , Tamoxifeno/uso terapêutico , Resultado do Tratamento
17.
Mol Cell Endocrinol ; 276(1-2): 63-70, 2007 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-17709176

RESUMO

Polymorphic variation of the LHR gene may affect receptor function and accordingly may influence ovarian steroid hormone action, including steroid hormone-dependent clinical outcome. The functional effects of two single nucleotide polymorphisms (SNPs), i.e. LHR 291Asn/Ser (rs12470652) and 312Ser/Asn (rs2293275) in the biologically interesting exon 10 of the LHR gene are described. Furthermore, ethnic diversity in allele frequencies and genotype distributions of both SNPs was determined. In addition associations with breast cancer were studied in 751 breast cancer patients. In vitro transfection studies revealed altered glycosylation status and increased receptor sensitivity for the 291Ser LHR variant. No functional consequences were observed for the 312SerAsn LHR SNP. The LHR 312Asn allele was slightly more often present in two independent breast cancer patient cohorts as compared to controls (OR=1.15; p=0.03 and 1.26; p=0.001, respectively). In conclusion, although functional changes of the LHR 291Ser candidate allele were observed, no associations with breast cancer were found, while the LHR 312Asn allele can be regarded as a weak breast cancer risk allele.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Éxons/genética , Predisposição Genética para Doença , Polimorfismo de Nucleotídeo Único/genética , Receptores do LH/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Alelos , Asparagina/genética , Estudos de Casos e Controles , Linhagem Celular , Feminino , Frequência do Gene , Genótipo , Humanos , Pessoa de Meia-Idade , Razão de Chances , Fatores de Risco , Serina/genética , Análise de Sobrevida
18.
Mol Cell Endocrinol ; 274(1-2): 30-4, 2007 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-17601657

RESUMO

Recent findings demonstrate that the effects of ghrelin can be abrogated by co-administered unacylated ghrelin (UAG). Since the general consensus is that UAG does not interact with the type 1a growth hormone secretagogue receptor (GHS-R), a possible mechanism of action for this antagonistic effect is via another receptor. However, functional antagonism of the GHS-R by UAG has not been explored extensively. In this study we used human GHS-R and aequorin expressing CHO-K1 cells to measure [Ca(2+)](i) following treatment with UAG. UAG at up to 10(-5)M did not antagonize ghrelin induced [Ca(2+)](i). However, UAG was found to be a full agonist of the GHS-R with an EC(50) of between 1.6 and 2 microM using this in vitro system. Correspondingly, UAG displaced radio-labeled ghrelin from the GHS-R with an IC(50) of 13 microM. In addition, GHS-R antagonists were found to block UAG induced [Ca(2+)](i) with approximately similar potency to their effect on ghrelin activation of the GHS-R, suggesting a similar mode of action. These findings demonstrate in a defined system that UAG does not antagonize activation of the GHS-R by ghrelin. But our findings also emphasize the importance of assessing the concentration of UAG used in both in vitro and in vivo experimental systems that are aimed at examining GHS-R independent effects. Where local concentrations of UAG may reach the high nanomolar to micromolar range, assignment of GHS-R independent effects should be made with caution.


Assuntos
Hormônios Peptídicos/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Animais , Células CHO , Cricetinae , Cricetulus , Grelina , Glucagon/metabolismo , Humanos , Hormônios Peptídicos/química , Peptídeos/metabolismo , Ensaio Radioligante , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Grelina , Somatostatina/metabolismo
19.
Hum Reprod ; 22(7): 1996-2005, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17582145

RESUMO

BACKGROUND: Ovarian failure as a complication of uterine artery embolization (UAE) for symptomatic uterine fibroids has raised concerns about this new treatment modality. METHODS: We investigated the occurrence of ovarian reserve reduction in a randomized trial comparing UAE and hysterectomy by measuring follicle stimulating hormone (FSH) and anti-Mullerian hormone (AMH). A total of 177 pre-menopausal women with menorrhagia due to uterine fibroids were included (UAE:n=88; hysterectomy:n=89). FSH and AMH were measured at baseline and at several time-points during the 24 months follow-up period. Follow-up AMH levels were also compared to the expected decrease due to ovarian ageing during the observational period. RESULTS: FSH increased significantly compared to baseline in both groups after 24 months follow-up (within group analysis: UAE:+12.1; P=0.001; hysterectomy:+16.3; P<0.0001). No differences in FSH values between the groups were found (P=0.32). At 24 months after treatment the number of patients with FSH levels>40 IU/l was 14/80 in the UAE group and 17/73 in the hysterectomy group (relative risk=0.75; P=0.37). AMH was measured in 63 patients (UAE: n=30; hysterectomy: n=33). After treatment AMH levels remained significantly decreased during the entire follow-up period only in the UAE group compared to the expected AMH decrease due to ageing. No differences were observed between the groups. CONCLUSIONS: This study shows that both UAE and hysterectomy affect ovarian reserve. This results in older women becoming menopausal after the intervention. Therefore, the application of UAE in women who still wish to conceive should only be considered after appropriate counselling.


Assuntos
Embolização Terapêutica/efeitos adversos , Hormônio Foliculoestimulante/sangue , Glicoproteínas/sangue , Histerectomia/efeitos adversos , Leiomioma/terapia , Doenças Ovarianas/etiologia , Hormônios Testiculares/sangue , Útero/irrigação sanguínea , Útero/patologia , Adulto , Envelhecimento , Hormônio Antimülleriano , Estrogênios/metabolismo , Feminino , Humanos , Menopausa , Pessoa de Meia-Idade , Ovário/patologia
20.
Am J Physiol Endocrinol Metab ; 293(3): E697-704, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17578884

RESUMO

Acylated and unacylated ghrelin (AG and UAG) are gut hormones that exert pleiotropic actions, including regulation of insulin secretion and glucose metabolism. In this study, we investigated whether AG and UAG differentially regulate portal and systemic insulin levels after a glucose load. We studied the effects of the administration of AG (30 nmol/kg), UAG (3 and 30 nmol/kg), the ghrelin receptor antagonist [D-Lys(3)]GHRP-6 (1 micromol/kg), or various combinations of these compounds on portal and systemic levels of glucose and insulin after an intravenous glucose tolerance test (IVGTT, d-glucose 1 g/kg) in anesthetized fasted Wistar rats. UAG administration potently and dose-dependently enhanced the rise of insulin concentration induced by IVGTT in the portal and, to a lesser extent, the systemic circulation. This UAG-induced effect was completely blocked by the coadministration of exogenous AG at equimolar concentrations. Similarly to UAG, [D-Lys(3)]GHRP-6, alone or in combination with AG and UAG, strongly enhanced the portal insulin response to IVGTT, whereas exogenous AG alone did not exert any further effect. Our data demonstrate that, in glucose-stimulated conditions, exogenous UAG acts as a potent insulin secretagogue, whereas endogenous AG exerts a maximal tonic inhibition on glucose-induced insulin release.


Assuntos
Glicemia/análise , Insulina/sangue , Oligopeptídeos/administração & dosagem , Hormônios Peptídicos/administração & dosagem , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Acetilação , Animais , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Grelina , Teste de Tolerância a Glucose , Masculino , Taxa de Depuração Metabólica/efeitos dos fármacos , Ratos , Ratos Wistar , Receptores de Grelina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA