Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Anim Reprod Sci ; 147(1-2): 56-63, 2014 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-24786546

RESUMO

The molecular mechanisms operating on a seasonal time-scale and regulating functions such as reproduction are poorly understood in animals living in desert environments. Kisspeptin, the product of the KiSS-1 gene, plays a critical role in control of the hypothalamic-pituitary-gonad axis that orchestrates the reproductive system in vertebrates. We report a sequence analysis of KiSS-1 and the pituitary luteinising hormone-beta (LHß) and follicle-stimulating hormone-beta (FSHß) in the Libyan jird (Meriones libycus), a seasonal breeding rodent that is sexually active during spring and quiescent in fall. We also assessed gene expression by quantitative real-time reverse transcription-polymerase chain reaction during the non-breeding and breeding seasons. The KiSS-1 cDNA sequence analysis showed high homology between M. libycus and all other rodents (94%) and humans (92%). KiSS-1 expression was higher during the breeding season than that during the non-breeding season. In contrast, LHß and FSHß expression levels were higher during the non-breeding season in autumn and varied in an opposite manner with testicular, seminal vesicle weights and plasma testosterone levels. Our results extend the role for KiSS-1 in activating the HPG axis in this desert rodent in its natural biotope by relaying environmental cues as in other seasonal non-desert rodent models.


Assuntos
Subunidade beta do Hormônio Folículoestimulante/metabolismo , Regulação da Expressão Gênica/fisiologia , Gerbillinae/metabolismo , Kisspeptinas/metabolismo , Hormônio Luteinizante Subunidade beta/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Subunidade beta do Hormônio Folículoestimulante/genética , Kisspeptinas/genética , Hormônio Luteinizante Subunidade beta/genética , Masculino , Dados de Sequência Molecular , Filogenia , Hipófise/fisiologia , Reprodução/fisiologia , Estações do Ano
2.
Adv Med Sci ; 57(2): 314-21, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23154428

RESUMO

PURPOSE: Loss-of-function mutations in FGFR1 have been identified in approximately 10% of the Kallmann syndrome (KS) patients. Previous reports have focused mainly on olfactory, reproductive, and some other features such as cleft lip/palate and dental agenesis. Given the ubiquitous expression of FGFR1 during development, other abnormal phenotypes might, however, have been overlooked in these patients. Here, we demonstrate skeletal phenotypic characterization of patients presented with KS and FGFR1 mutations. MATERIAL AND METHODS: Using the Sanger DNA sequencing technique a cohort of 29 KS patients was screened. RESULTS: Here, we report on 5 KS patients who carry FGFR1 mutations (Gly270Asp, Gly97Ser, Met161Thr, Ser685Phe and Ala167Ser/Ala167Ser). Three patients presented with skeletal abnormalities, i.e. spine (hemivertebra and butterfly vertebra) and limb (oligodactyly of the feet, fusion of the 4th and 5th metacarpal bones) malformations in two patients and one patient, respectively. The hand phenotype found in the patient cannot be thought of as a counter-type of the hand phenotype resulting from FGFR1 gain-of-function mutations. The skeletal anomalies identified in the 3 KS patients are close to those observed in Fgfr1 conditional knockout mice. CONCLUSIONS: This study demonstrates that FGFR1 loss-of-function mutations can be associated with skeletal abnormalities also in humans. Further investigations in KS patients who carry FGFR1 mutations are needed to evaluate the prevalence of skeletal defects in this genetic form of KS. Conversely, the presence of bone malformations in a KS patient should direct the geneticist towards a search for mutations in FGFR1.


Assuntos
Osso e Ossos/anormalidades , Síndrome de Kallmann/genética , Síndrome de Kallmann/patologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Adolescente , Adulto , Sequência de Aminoácidos , Substituição de Aminoácidos , Desenvolvimento Ósseo/genética , Feminino , Humanos , Masculino , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Fenótipo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/química , Adulto Jovem
3.
J Clin Endocrinol Metab ; 95(6): 3028-38, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20427508

RESUMO

CONTEXT: Heterozygous GNAS inactivating mutations are known to induce pseudohypoparathyroidism type 1a when maternally inherited and pseudopseudohypoparathyroidism when paternally inherited. Progressive osseous heteroplasia (POH) is a rare disease of ectopic bone formation, and studies in different families have shown that POH is also caused by paternally inherited GNAS mutations. OBJECTIVE: Our purpose was to characterize parental origin of the mutated allele in de novo cases of POH and to draw phenotype/genotype correlations according to maternal or paternal transmission of a same GNAS mutation. DESIGN AND SETTING: We conducted a retrospective study on patients addressed to our referral center for the rare diseases of calcium and phosphorus metabolism. PATIENTS AND METHODS: We matched 10 cases of POH with cases of pseudohypoparathyroidism type 1a carrying the same GNAS mutations. MAIN OUTCOME MEASURES: The parental origin of the mutated allele was studied using informative intragenic polymorphisms and subcloning of PCR products. RESULTS: Paternal origin of GNAS mutations was clearly demonstrated in eight POH cases including one patient with mutation in exon 1. Genotype/phenotype analyses suggest that there is no direct correlation between the ossifying process and the position of the inactivating GNAS mutation. It is, however, more severe in patients in whom origin of the mutation is paternal. Severe intrauterine growth retardation was clearly evidenced in paternally inherited mutations. CONCLUSIONS: Clinical heterogeneity makes genetic counseling a delicate matter, especially in which paternal inheritance is concerned because it can lead to either a mild expression of pseudopseudohypoparathyroidism or a severe expression of POH.


Assuntos
Osso e Ossos , Coristoma/genética , Coristoma/patologia , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Mutação/genética , Mutação/fisiologia , Criança , Pré-Escolar , Cromograninas , Metilação de DNA , Bases de Dados Genéticas , Feminino , Impressão Genômica , Genótipo , Humanos , Lactente , Masculino , Hormônio Paratireóideo/fisiologia , Linhagem , Polimorfismo de Nucleotídeo Único , Pseudo-Hipoparatireoidismo/genética , Pseudopseudo-Hipoparatireoidismo/genética , RNA/genética
4.
Pathol Biol (Paris) ; 58(5): 367-71, 2010 Oct.
Artigo em Francês | MEDLINE | ID: mdl-19942373

RESUMO

Parental imprinting and the type of the genetic alteration play a determinant role in the phenotype expression of GNAS locus associated to pseudohypoparathyroidism (PHP). This imprint is tissue-specific, mainly localized in the kidney and the thyroid. Only the maternal allele is expressed at this level. An alteration in the coding sequence of the gene leads to an haplo-insufficiency and a dysmorphic phenotype (Albright's syndrome). If the alteration is on the maternal allele, there is a hormonal resistance to the PTH at the kidney level and to the TSH at the thyroid level. The phenotype is known as a PHP1a. If the alteration is on the paternal allele, there are few clinical signs with no hormonal resistance and the phenotype is known as pseudo-pseudo-hypoparathyroidism (PPHP). Methylation anomalies of GNAS locus, in particular of exon 1A, are responsible for a lack of expression of Gαs at kidney and thyroid levels only. If these anomalies concern the maternal allele (the only one expressed) with a paternal pattern, there is no haplo-insufficiency and no dysmorphic syndrome. The hormonal resistance is yet again limited to PTH and TSH. The phenotype is known as PHP1b. In the familial forms, these methylation anomalies are associated with a deletion of the syntaxine 16 gene in the maternal allele. This gene contains probably the imprinting center of the locus.


Assuntos
Epigênese Genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Impressão Genômica/genética , Pseudo-Hipoparatireoidismo/genética , Animais , Cromograninas , Metilação de DNA , Feminino , Displasia Fibrosa Poliostótica/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/deficiência , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Genes Dominantes , Humanos , Rim/metabolismo , Masculino , Camundongos , Camundongos Knockout , Especificidade de Órgãos , Hormônio Paratireóideo/fisiologia , Fenótipo , Pseudopseudo-Hipoparatireoidismo/genética , Sintaxina 16/genética , Sintaxina 16/fisiologia , Glândula Tireoide/metabolismo , Tireotropina/fisiologia
6.
Folia Histochem Cytobiol ; 47(5): S55-8, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-20067895

RESUMO

We report a woman with primary amenorrhoea and infertility associated with an isolated deficiency of pituitary FSH that does not respond to GnRH administration. Serum inhibin B was undetectable and antimullerian hormone (AMH) was within the normal range. Ultra sound examination revealed a small uterus and small ovaries with few small follicles. We identified an homozygous 1-bp (G) deletion at codon 79 in FSHbeta gene suggesting a complete loss of function. The patient underwent studies of ovarian responsiveness to recombinant human FSH according to the following protocol: 150UI/d for five days following by 75 UI/d for 10 days. Estradiol plasma level started to increase from day 5 associated to a sharp increase of inhibine B and a decrease of LH. During the same time, we observed an excessive development of multiple follicles resulting in an arrest of the treatment to avoid hyperstimulation. The present study confirm that follicles up to 5 mm in diameter had developed in the absence of FSH and that FSH is required for the growth of follicles beyond the two-layer granulose stage.


Assuntos
Subunidade beta do Hormônio Folículoestimulante , Hormônio Foliculoestimulante , Estradiol/sangue , Feminino , Hormônio Foliculoestimulante/sangue , Hormônio Foliculoestimulante Humano , Humanos , Hipogonadismo , Hormônio Luteinizante/sangue , Mutação , Folículo Ovariano , Puberdade Tardia
7.
J Neuroendocrinol ; 20(3): 381-93, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18208554

RESUMO

Kisspeptin, the product derived from KiSS-1, and its cognate receptor, GPR54, both exert a role in the neuroendocrine control of reproduction by regulating gonadotrophin-releasing hormone (GnRH) secretion. In the present study, we demonstrate, using dual immunofluorescence with specific antibodies, that the KiSS-1 and GPR54 genes are both expressed in rat gonadotrophs. All luteinising hormone beta-immunoreactive (LH beta-ir) cells were stained by the KiSS-1 antibody but some kisspeptin-ir cells were not LH beta positive; thus, we cannot exclude the possibility that kisspeptins are expressed in other pituitary cells. All GPR54-ir are co-localised with LH beta cells, but only a subset of LH beta cells are stained with the GPR54 antibody. Using the real-time reverse transcription-polymerase chain reaction (RT-PCR), we found that the expression of KiSS-1 and GPR54 is differentially regulated by steroids. In the female, KiSS-1 mRNA levels dramatically decreased following ovariectomy (OVX), and this decrease was prevented by administration of 17beta-oestradiol (E(2)), but not by administration of GnRH antagonist or agonist. Administration of E(2) in OVX rats receiving either GnRH antagonist or agonist clearly shows that E(2) acts directly on the pituitary to positively control KiSS-1 expression. In OVX rats, administration of the selective oestrogen receptor (ER)alpha ligand propylpyrazoletriol, but not the selective ER beta ligand diarylpropionitrile, mimics this effect. By contrast, our study shows that GPR54 expression is positively regulated by GnRH and negatively controlled by chronic exposure to E(2). In summary, our data document for the first time that, in the female rat pituitary, KiSS-1 expression is up-regulated by oestradiol, similarly to that seen in the anteroventral periventricular nucleus of the hypothalamus. Conversely, GPR54 is up-regulated by GnRH, which exclusively targets gonadotrophs.


Assuntos
Estradiol/fisiologia , Regulação da Expressão Gênica , Gonadotrofos/metabolismo , Hormônio Liberador de Gonadotropina/fisiologia , Proteínas/genética , Receptores Acoplados a Proteínas G/genética , Animais , Células Cultivadas , Estradiol/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Kisspeptinas , Masculino , Hipófise/metabolismo , Proteínas/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Estrogênio/metabolismo , Receptores de Estrogênio/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Kisspeptina-1
9.
J Clin Endocrinol Metab ; 86(8): 3746-53, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11502806

RESUMO

Familial medullary thyroid carcinoma only is related to germline mutations in the protooncogene RET, mainly in exons 10, whereas noncysteine mutations (exons 13-15) are considered infrequent. We analyzed 148 patients from 47 familial medullary thyroid carcinoma only families, and we found noncysteine RET mutations in 59.5% of these families. Of the index cases with noncysteine mutations, 43.4% presented with a multinodular goiter and high basal calcitonin; they were older at diagnosis than those with mutation in exon 10 and had more multifocal medullary thyroid carcinoma, but no difference in size, bilaterality, presence of C cell hyperplasia, or nodal metastases was found. Gene carriers with noncysteine RET mutations had a lower incidence of medullary thyroid carcinoma (78.2% vs. 94.1%) than those with mutation in exon 10; 20.2% had C cell hyperplasia only, although thyroidectomized at an older age. In conclusion, familial medullary thyroid carcinoma with noncysteine RET mutations are not infrequent and are overrepresented in presumed sporadic medullary thyroid carcinoma, suggesting that RET analysis should routinely be extended to exons 13, 14, and 15. The phenotype is characterized by a late onset of the disease, suggesting a delayed appearance of C cell disease rather than a less aggressive form. In familial medullary thyroid carcinoma gene carriers, the optimal timing for thyroidectomy remains controversial. Based on these data, we propose that surgery should be performed before elevation of the basal calcitonin level, potentially as soon as the pentagastrin test becomes abnormal.


Assuntos
Carcinoma Medular/genética , Proteínas de Drosophila , Proteínas Proto-Oncogênicas/genética , Proto-Oncogenes , Receptores Proteína Tirosina Quinases/genética , Neoplasias da Glândula Tireoide/genética , Adulto , Calcitonina/sangue , Carcinoma Medular/epidemiologia , Carcinoma Medular/patologia , Carcinoma Medular/cirurgia , Cisteína , Bases de Dados como Assunto , Éxons , Feminino , França , Triagem de Portadores Genéticos , Ligação Genética , Genótipo , Humanos , Hiperplasia , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Pentagastrina , Fenótipo , Proteínas Proto-Oncogênicas c-ret , Glândula Tireoide/patologia , Neoplasias da Glândula Tireoide/epidemiologia , Neoplasias da Glândula Tireoide/patologia , Neoplasias da Glândula Tireoide/cirurgia , Tireoidectomia
10.
Am J Med Genet ; 99(2): 132-6, 2001 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-11241472

RESUMO

Mazabraud syndrome is a rare sporadic disorder, mainly characterized by bone fibrous dysplasia and intramuscular myxomas. We report here two new cases of Mazabraud syndrome. One of our patients (Patient 1) also had café-au-lait spots and multinodular goiter suggestive of McCune-Albright syndrome. We review the 37 previously reported cases with Mazabraud syndrome and discuss the 6/37 patients with criteria of Mazabraud and McCune-Albright syndromes. Based on the clinical overlap between the two syndromes, we tested the GNAS1 gene in blood leukocytes and skin fibroblasts of Patient 1, but found no evidence of an activating mutation in the GNAS1 gene.


Assuntos
Anormalidades Múltiplas , Displasia Fibrosa Poliostótica , Anormalidades Múltiplas/diagnóstico por imagem , Anormalidades Múltiplas/genética , Adulto , Feminino , Displasia Fibrosa Poliostótica/diagnóstico por imagem , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Humanos , Neoplasias Musculares , Mixoma , Radiografia , Síndrome
11.
J Clin Endocrinol Metab ; 85(9): 3002-8, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10999776

RESUMO

We describe a woman with complete hypogonadotropic hypogonadism and a new compound heterozygous mutation of the GnRH receptor (GnRHR) gene. A null mutation L314X leading to a partial deletion of the seventh transmembrane domain of the GnRHR is associated with a Q106R mutation previously described. L314X mutant receptor shows neither measurable binding nor inositol phosphate production when transfected in CHO-K1 cells compared to the wild-type receptor. The disease is transmitted as an autosomal recessive trait, as shown by pedigree analysis. Heterozygous patients with GnRHR mutations had normal pubertal development and fertility. The present study shows an absence of LH and FSH response to pulsatile GnRH administration (20 microg/pulse, sc, every 90 min). However, GnRH triggered free alpha-subunit (FAS) pulses of small amplitude, demonstrating partial resistance to pharmacological doses of GnRH. FSH, LH, and FAS concentrations were evaluated under chronic estrogen treatment and repeat administration of GnRH. Not only were plasma FSH, LH, and FAS concentrations decreased, but FAS responsiveness was reduced. This new case emphasizes the implication of the GnRH receptor mutations in the etiology of idiopathic hypogonadotropic hypogonadism. We also have evidence for a direct negative estrogen effect on gonadotropin secretion at the pituitary level, dependent on the GnRHR signaling pathway.


Assuntos
Estrogênios/efeitos adversos , Gonadotropinas/deficiência , Hipogonadismo/genética , Mutação/genética , Receptores LHRH/genética , Sequência de Aminoácidos , Animais , Células CHO , Criança , Cricetinae , Feminino , Hormônio Foliculoestimulante/sangue , Subunidade alfa de Hormônios Glicoproteicos/sangue , Gonadotropinas/farmacologia , Haplótipos , Humanos , Hormônio Luteinizante/sangue , Dados de Sequência Molecular , Fenótipo , Conformação Proteica , RNA Mensageiro/biossíntese , RNA Mensageiro/genética
12.
J Endocrinol ; 162(3): 409-15, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10467232

RESUMO

The demonstration of an inhibitory effect of gonadotropin-releasing hormone (GnRH) agonists upon steroidogenesis in hypophysectomized rats and the presence of mRNA coding for GnRH and GnRH receptors (GnRH-R) in rat gonads suggests that GnRH can act locally in the gonads. To assess this hypothesis, we investigated the effects of GnRH analogs, gonadotropins and testosterone on the levels of both GnRH and GnRH-R mRNA in the rat testis. Using dot blot hybridization, we measured the mRNA levels 2 to 120 h after the administration of the GnRH agonist, triptorelin. We observed an acute reduction of both GnRH and GnRH-R mRNAs 24 h after the injection (about 38% of control). However, the kinetics for testis GnRH-R mRNA were different from those previously found for pituitary GnRH-R mRNA under the same conditions. Initially, the concentrations of serum LH and FSH peaked, then declined, probably due to the desensitization of the gonadotrope cells. In contrast, the GnRH antagonist, antarelix, after 8 h induced a 2.5-fold increase in GnRH-R mRNA, but not in GnRH mRNA, while gonadotropins levels were reduced. Human recombinant FSH had no significant effect on either GnRH or GnRH-R mRNA levels. Inversely, GnRH-R mRNA levels markedly decreased by 21% of that of control 24 h after hCG injection. Finally, 24 h after testosterone injection, a significant increase in GnRH-R mRNA levels (2.3 fold vs control) was found, but a reduction in the concentration of serum LH, probably by negative feedback on the pituitary, was observed. In contrast, GnRH mRNA levels were not significantly altered following testosterone treatment. Since LH receptors, GnRH-R and testosterone synthesis are colocalized in Leydig cells, our data suggest that LH could inhibit the GnRH-R gene expression or decrease the GnRH-R mRNA stability in the testis. However, this does not exclude the possibility that GnRH analogs could also affect the GnRH-R mRNA levels via direct binding to testicular GnRH-R. In contrast, the regulation of GnRH mRNA levels appeared to be independent of gonadotropins. Taken together, our results suggest a regulation of GnRH and GnRH-R mRNA specific for the testis.


Assuntos
Hormônio Liberador de Gonadotropina/genética , Hormônio Luteinizante/farmacologia , RNA Mensageiro/metabolismo , Receptores LHRH/metabolismo , Testículo/metabolismo , Análise de Variância , Animais , Gonadotropina Coriônica/farmacologia , Hormônio Liberador de Gonadotropina/agonistas , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Immunoblotting , Masculino , Oligopeptídeos/farmacologia , RNA Mensageiro/análise , Ratos , Ratos Wistar , Receptores LHRH/genética , Testículo/efeitos dos fármacos , Testosterona/farmacologia , Pamoato de Triptorrelina/farmacologia
13.
Eur J Endocrinol ; 140(6): 561-9, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10366411

RESUMO

The expression pattern of the GnRH receptor was investigated in a variety of normal and neoplastic human tissues by RT-PCR-Southern blotting. In addition to the full-length cDNA (sb1), we identified two other transcripts: the first (sb2) was characterized by a 128 bp deletion as previously described; the second was an unexpected finding composed of a shorter cDNA (sb3), the sequence of which revealed a 220 bp deletion corresponding in size to exon 2. These three transcripts were found in normal pituitary and pituitary adenomas, and in granulosa tumors, but not in testis, where sb2 was lacking. Only sb1 was expressed in normal, fibrocystic and malignant breast tissue. No transcript with a full-length region was found in endometrium, intestine or lymphocytes. This is the first report that shows that splicing of the gonadotropin-releasing hormone receptor gene is tissue dependent. We also determined the intron-exon nucleotide sequence of the gene and identified an MaeIII polymorphic site in exon 1 created by a silent C453T transition found in 10% of unrelated French whites.


Assuntos
Receptores LHRH/genética , Adenoma/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Clonagem Molecular , DNA Complementar/análise , Regulação da Expressão Gênica , Humanos , Masculino , Dados de Sequência Molecular , Mutação , Hipófise/metabolismo , Neoplasias Hipofisárias/metabolismo , Splicing de RNA , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Testículo/metabolismo
14.
J Clin Endocrinol Metab ; 84(3): 990-6, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10084584

RESUMO

We have studied a kindred with three siblings with isolated hypogonadotropic hypogonadism caused by compound heterozygote mutations in the GnRH receptor gene. The disorder was transmitted as an autosomal recessive trait. The R262Q mutation in intracellular loop 3 of the receptor was associated with a mutation in the third transmembrane domain of the receptor, A129D, that has never been described before. This A129D mutation results in a complete loss of function, indicated by the lack of inositol triphosphate (TP3) 3 production by transfected Chinese hamster ovary (CHO) cells after GnRH stimulation. The two brothers had microphallus and bilateral cryptorchidism and were referred for lack of puberty, whereas their sister had primary amenorrhea and a complete lack of puberty. Their basal gonadotropin concentrations were below the reference range, and their endogenous LH secretory patterns were abnormal, with a low-normal frequency of small pulses or no apparent LH pulse. Pulsatile GnRH administration (10 microg/pulse every 90 min for 40 h) resulted in increased mean LH without any significant changes in testosterone levels in the two brothers, whereas the LH secretory profile of their sister remained apulsatile. Larger pulses of exogenous GnRH (20 microg every 90 min for 24 h) caused the sister to produce recognizable low amplitude LH pulses. The concentrations of free alpha-subunit significantly increased in all patients during the pulsatile GnRH administration. Thus, these hypogonadal patients are partially resistant to pulsatile GnRH administration, suggesting that they should be treated with gonadotropins to induce spermatogenesis or ovulation rather than with pulsatile GnRH.


Assuntos
Hormônio Liberador de Gonadotropina/uso terapêutico , Hipogonadismo/tratamento farmacológico , Hipogonadismo/genética , Mutação/fisiologia , Receptores LHRH/genética , Adolescente , Adulto , Sequência de Aminoácidos/genética , Animais , Células CHO , Cricetinae , DNA/genética , Resistência a Medicamentos/fisiologia , Feminino , Haplótipos , Humanos , Hormônio Luteinizante/metabolismo , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Fluxo Pulsátil
15.
Arch Med Res ; 30(6): 481-5, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10714361

RESUMO

Mutations in a few genes have been identified in hypogonadotropic hypogonadism (HH): the gene KAL-1 is involved in X-linked Kallmann syndrome associated with anosmia and mutations in transcription factors, namely, DAX-1 and Prop-1 were also evidenced when associated with other pituitary or endocrine defects. Recently, compound heterozygote mutations in the GnRH receptor gene were described both in males and females and hormonal resistance was confirmed in vitro. There is a wide spectrum of phenotype, ranging from complete HH with lack of pubertal development and cryptorchidism to partial hypogonadism with an arrest of pubertal development. In complete GnRH resistance, endogenous LH secretory patterns were abnormal, either apulsatile or characterized by a low-normal pulse frequency with small pulses or erratic pulses of low amplitude. In patients with partial resistance, basal LH plasma concentration was low, but FSH level was in the normal range. LH pulse analysis revealed normal frequency with decreased amplitude. Mutations are distributed along the coding sequence, as reported for other GPCRs. However, two hot-spots, Q106R and the R262Q, were observed, regardless of the geographic origin of the patients. In most cases, patients responded to GnRH administration, making the GnRH test inappropriate for screening GnRH resistance in IHH.


Assuntos
Genes Recessivos , Hormônio Liberador de Gonadotropina/deficiência , Hipogonadismo/genética , Mutação , Receptores LHRH/genética , Sequência de Aminoácidos , Animais , Humanos , Hipogonadismo/metabolismo , Dados de Sequência Molecular
16.
Clin Endocrinol (Oxf) ; 49(1): 115-23, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9797855

RESUMO

OBJECTIVE: Given the central role of the GnRH receptor (GnRHR) in the regulation of the gonadotrophin secretion, it might be implicated directly or indirectly in the pathogenesis of gonadotroph tumours. DESIGN: We determined if GnRHR mRNA was expressed in gonadotroph tumours using RT-PCR and analysed the GnRHR gene for the presence of mutations in its coding region, using direct sequencing of PCR products. Results were analysed according to the pattern of expression of alpha, beta-FSH and beta-LH subunit (SU) genes. SUBJECTS: RNA was extracted from 20 gonadotroph tumours identified by immunohistochemistry (> 10% of stained cells): 9 adenomas were functioning (high serum gonadotrophin levels), 3 were associated with high alpha-SU levels and 8 were nonfunctioning. Genomic DNA was extracted from 64 normal subjects. RESULTS: We found GnRHR mRNA in 12 tumours (60%): 8/9 functioning (88%), 1/3 alpha-secreting (33%) and 3/8 nonfunctioning (37.5%) gonadotroph adenomas. There was a significant association between GnRHR expression and immunostaining for beta-FSH (P = 0.014). The nucleotide sequence of the amplified products was identical to that of human pituitary except for the presence, in 3 functioning adenomas, of a silent C to T transition at nucleotide 453 encoding for the serine residue situated in the second intracellular loop at position 151. Heterozygosity provided evidence that both alleles were transcribed in these tumours. This substitution creates a Mae III restriction site. Genomic DNA from normal subjects were then tested for the presence of this new polymorphism. The frequency of the heterozygosity (18.7%) was not significantly different from that found in gonadotroph tumours (25%) and this new Mae III polymorphism site cannot be used as a tumoural marker. CONCLUSION: The GnRHR gene is preferentially expressed in functioning rather than in nonfunctioning gonadotroph adenomas, but no mutations altering the coding region of the gene were found to further substantiate its role in the pathogenesis of gonadotroph tumours.


Assuntos
Adenoma/genética , Adenoma/metabolismo , Gonadotropinas Hipofisárias/metabolismo , Neoplasias Hipofisárias/genética , Neoplasias Hipofisárias/metabolismo , Receptores LHRH/genética , Adolescente , Adulto , Idoso , Sequência de Bases , Feminino , Expressão Gênica , Heterozigoto , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Polimorfismo Genético
17.
J Endocrinol ; 159(1): 179-89, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9795356

RESUMO

The identification of gonadal gonadotropin-releasing hormone receptors (GnRH-R) and evidence of direct inhibitory effects of GnRH agonists upon steroidogenesis in adult rat gonads, lend credence to a putative intragonadal role of a locally secreted GnRH or GnRH-like peptide. Using reverse transcription-polymerase chain reaction followed by Southern blot hybridization and sequencing, we identified, both in the ovary and in the testis of fetal and adult rats, a fully processed GnRH messenger RNA (mRNA), the sequence of which, in adult testis, was identical to that found in the hypothalamus. We also detected in the testis, but not in the ovary, a transcript containing the first intron. The ontogeny of GnRH and GnRH-R gene expression was studied in rat gonads from 14.5 to 21.5 days post-coitum (dpc), using dot blot hybridization of total RNA. During this period, the levels of cyclophilin mRNA normalized to total RNA remained unchanged. Thus, we used cyclophilin as an internal standard. GnRH mRNA was detected in the ovary at 18.5 dpc, four days later than in the testis, and similar levels were found in both sexes at birth. GnRH-R mRNA was present at 14.5 dpc in the testis and at 15.5 dpc in the ovary, with the levels at 21.5 dpc being 2.4 times higher in the testis than in the ovary. GnRH and GnRH-R mRNA levels increased in both sexes in late fetal development, but this increase appeared two days sooner in the ovary compared with the testis, thus supporting the hypothesis that expression of the GnRH and GnRH-R genes is regulated in a sex-dependent manner during fetal development. In all cases, expression of GnRH and GnRH-R preceded gonadotropin receptors in the gonads and initiation of gonadotropin secretion by the pituitary.


Assuntos
Desenvolvimento Embrionário e Fetal/genética , Hormônio Liberador de Gonadotropina/genética , Ovário/embriologia , Receptores LHRH/genética , Testículo/embriologia , Animais , Southern Blotting , Densitometria , Eletroforese em Gel de Ágar , Feminino , Expressão Gênica , Masculino , Ovário/metabolismo , Peptidilprolil Isomerase/genética , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Testículo/metabolismo
18.
J Clin Endocrinol Metab ; 83(10): 3450-3, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9768644

RESUMO

We report an unusual case of a gonadotroph adenoma in a 34-yr-old woman, revealed by a dramatic rise in the plasma estradiol (E2) concentration (26,800 pmol/L; normal, <370), with nonsuppressed FSH and LH levels (4.9 and 2.4 mIU/mL, respectively). The PRL level was 503 ng/mL. The testosterone and progesterone levels were 7 and 17 nmol/L, respectively. The levels of inhibin alpha, inhibin A, and inhibin B were increased compared to normal values in both the follicular (fp) and luteal (lp) phases of the menstrual cycle [inhibin alpha, 1986 IU/L (fp normal, <700; lp normal, <1650); inhibin A, 254 pg/mL (fp normal, <20; lp normal, <120); inhibin B, 246 pg/mL (fp normal, <150; lp normal, <30 lp)]. Pituitary magnetic resonance imaging revealed a huge pituitary adenoma. After transphenoidal surgery, the patient presented with pituitary insufficiency and diabetes insipidus. RT-PCR of the tumor tissue was positive for LHbeta, FSHbeta, alpha-subunit, and PRL. This case is of particular interest because 1) although the E2 level was extremely high, the patient did not present with ascitis, suggesting that chronic elevated E2 does not play a crucial role in the hyperstimulation symptoms; 2) the extreme rise in E2 was related to the cosecretion of FSH and LH, confirming the two-cell two-gonadotropin theory; and 3) the rise in inhibin B is associated with FSH secretion, whereas the rise in inhibin A is probably due to luteinization.


Assuntos
Adenoma/diagnóstico , Adenoma/metabolismo , Hormônio Foliculoestimulante/metabolismo , Hormônio Luteinizante/metabolismo , Síndrome de Hiperestimulação Ovariana/diagnóstico , Neoplasias Hipofisárias/diagnóstico , Neoplasias Hipofisárias/metabolismo , Adenoma/sangue , Adenoma/cirurgia , Adulto , Idoso , Diagnóstico Diferencial , Feminino , Hormônio Foliculoestimulante/sangue , Hormônios/sangue , Humanos , Hormônio Luteinizante/sangue , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Neoplasias Hipofisárias/sangue , Neoplasias Hipofisárias/cirurgia
19.
Int J Cancer ; 71(4): 595-9, 1997 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-9178813

RESUMO

While gonadotropin-releasing hormone (GnRH) or GnRH receptor (GnRHR) have been reported to exist in tissues other than brain and pituitary, there is no report concerning co-expression of GnRH and GnRHR in human breast tissues. To address this question, we have examined whether mRNA for GnRH as well as GnRHR was present in different human breast samples, by employing the reverse transcription-polymerase chain reaction (RT-PCR) protocol followed by Southern blotting of the PCR products. Coexpression of GnRH and GnRHR genes was further confirmed by dot blot hybridization using appropriate [32P]-labeled probes. We thus tested fibrocystic breast (4 cases), invasive ductal carcinomas (6 cases) and 1 adjacent non-neoplastic tissue. GnRHR and GnRH mRNAs were found in all actin-positive samples including malignant as well as nonmalignant tissues. Quantitative determinations of mRNA did not reveal significant differences between malignant and non-malignant breast samples for either GnRH or GnRHR gene expression. Our data show that neither gene was overexpressed in the breast cancer samples compared with normal breast tissue. Since GnRH agonists inhibit breast epithelial cell growth, the presence of GnRHR mRNA suggests that GnRH may specifically affect breast cell growth. Our data thus raise the possibility of an autocrine/paracrine role for GnRH in human mammary gland.


Assuntos
Neoplasias da Mama/metabolismo , Doença da Mama Fibrocística/metabolismo , Hormônio Liberador de Gonadotropina/biossíntese , Proteínas de Neoplasias/biossíntese , Receptores LHRH/biossíntese , Mama/metabolismo , Neoplasias da Mama/genética , Transformação Celular Neoplásica/genética , DNA Complementar/genética , DNA de Neoplasias/genética , Feminino , Doença da Mama Fibrocística/genética , Regulação Neoplásica da Expressão Gênica , Hormônio Liberador de Gonadotropina/genética , Humanos , Proteínas de Neoplasias/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Neoplásico/biossíntese , RNA Neoplásico/genética , Receptores LHRH/genética
20.
Hum Reprod ; 10(11): 2904-8, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8747041

RESUMO

The prognostic value of the oestradiol flare-up in response to gonadotrophin-releasing hormone (GnRH) agonist was evaluated in 140 in-vitro fertilization (IVF) cycles programmed by progestogen pretreatment. Three days after the end of administration of norethisterone, a routinely used short-term DTRP6 GnRH agonist protocol was started (designated day 1), gonadotrophins being introduced from day 4. Serum oestradiol flare-up values were evaluated on days 1, 2 and 3 to study their relationship with the subsequent IVF outcome. On day 2, 87.9% of the cycles exhibited a significant rise in serum oestradiol concentration from baseline (delta E2 > or = 5 pg/ml). Compared to cycles without any significant oestradiol increase, they had a higher pregnancy rate per transfer (33.3 versus 9.1%, P = 0.02), although the number of transferred embryos did not differ significantly. Taking into account the previously described cut-off value (doubling from baseline), we found that less than half of the cycles (45.7%) involved a doubling of oestradiol values during flare-up, and we did not observe any significant difference in IVF outcome in these cycles compared to those without doubling. In conclusion, progestogen pretreatment, by inducing ovarian quiescence, may lower the oestradiol cut-off value that is predictive of the subsequent pregnancy rate. Nevertheless, determination of the absolute oestradiol response (delta E2) to GnRH agonist after progestogen pretreatment could allow a further adaptation of the protocol to achieve an optimum response in each cycle. Another alternative for patients with a lower delta E2 could be the suppression of progestogen pretreatment.


Assuntos
Estradiol/sangue , Hormônio Liberador de Gonadotropina/agonistas , Noretindrona/administração & dosagem , Indução da Ovulação/métodos , Congêneres da Progesterona/administração & dosagem , Pamoato de Triptorrelina/administração & dosagem , Adulto , Protocolos Clínicos , Feminino , Fertilização in vitro , Humanos , Infertilidade/sangue , Infertilidade/terapia , Luteolíticos/administração & dosagem , Masculino , Gravidez , Prognóstico , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA