Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
PLoS One ; 11(7): e0160292, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27467252

RESUMO

A gonadotropin-releasing hormone (GnRH)-like molecule was previously identified in a gastropod, Aplysia californica, and named ap-GnRH. In this study, we cloned the full-length cDNA of a putative ap-GnRH receptor (ap-GnRHR) and functionally authenticated this receptor as a bona fide ap-GnRHR. This receptor contains two potential translation start sites, each accompanied by a Kozak sequence, suggesting the translation of a long and a short form of the receptor is possible. The putative ap-GnRHR maintains the conserved structural motifs of GnRHR-like receptors and shares 45% sequence identity with the octopus GnRHR. The expression of the putative ap-GnRHR short form is ubiquitous in all tissues examined, whereas the long form is only expressed in parts of the central nervous system, osphradium, small hermaphroditic duct, and ovotestis. The cDNA encoding the long or the short receptor was transfected into the Drosophila S2 cell line and subject to a radioreceptor assay using 125I-labeled ap-GnRH as the radioligand. Further, the transfected cells were treated with various concentrations of ap-GnRH and measured for the accumulation of cAMP and inositol monophosphate (IP1). Radioreceptor assay revealed that only the long receptor bound specifically to the radioligand. Further, only the long receptor responded to ap-GnRH with an increased accumulation of IP1, but not cAMP. Our studies show that despite the more prevalent expression of the short receptor, only the long receptor is the functional ap-GnRHR. Importantly, this is only the second report on the authentication of a protostome GnRHR, and based on the function and the phylogenetic grouping of ap-GnRHR, we suggest that this receptor is more similar to protostome corazonin receptors than chordate GnRHRs.


Assuntos
Evolução Biológica , Receptores LHRH/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Células Cultivadas , Clonagem Molecular , DNA Complementar/genética , Gastrópodes , Filogenia , Ensaio Radioligante , Receptores LHRH/genética , Receptores LHRH/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos
2.
Hum Reprod Update ; 22(3): 358-81, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26715597

RESUMO

BACKGROUND: Human reproduction depends on an intact hypothalamic-pituitary-gonadal (HPG) axis. Hypothalamic gonadotrophin-releasing hormone (GnRH) has been recognized, since its identification in 1971, as the central regulator of the production and release of the pituitary gonadotrophins that, in turn, regulate the gonadal functions and the production of sex steroids. The characteristic peculiar development, distribution and episodic activity of GnRH-producing neurons have solicited an interdisciplinary interest on the etiopathogenesis of several reproductive diseases. The more recent identification of a GnRH/GnRH receptor (GnRHR) system in both the human endometrium and ovary has widened the spectrum of action of the peptide and of its analogues beyond its hypothalamic function. METHODS: An analysis of research and review articles published in international journals until June 2015 has been carried out to comprehensively summarize both the well established and the most recent knowledge on the physiopathology of the GnRH system in the central and peripheral control of female reproductive functions and diseases. RESULTS: This review focuses on the role of GnRH neurons in the control of the reproductive axis. New knowledge is accumulating on the genetic programme that drives GnRH neuron development to ameliorate the diagnosis and treatment of GnRH deficiency and consequent delayed or absent puberty. Moreover, a better understanding of the mechanisms controlling the episodic release of GnRH during the onset of puberty and the ovulatory cycle has enabled the pharmacological use of GnRH itself or its synthetic analogues (agonists and antagonists) to either stimulate or to block the gonadotrophin secretion and modulate the functions of the reproductive axis in several reproductive diseases and in assisted reproduction technology. Several inputs from other neuronal populations, as well as metabolic, somatic and age-related signals, may greatly affect the functions of the GnRH pulse generator during the female lifespan; their modulation may offer new possible strategies for diagnostic and therapeutic interventions. A GnRH/GnRHR system is also expressed in female reproductive tissues (e.g. endometrium and ovary), both in normal and pathological conditions. The expression of this system in the human endometrium and ovary supports its physiological regulatory role in the processes of trophoblast invasion of the maternal endometrium and embryo implantation as well as of follicular development and corpus luteum functions. The GnRH/GnRHR system that is expressed in diseased tissues of the female reproductive tract (both benign and malignant) is at present considered an effective molecular target for the development of novel therapeutic approaches for these pathologies. GnRH agonists are also considered as a promising therapeutic approach to counteract ovarian failure in young female patients undergoing chemotherapy. CONCLUSIONS: Increasing knowledge about the regulation of GnRH pulsatile release, as well as the therapeutic use of its analogues, offers interesting new perspectives in the diagnosis, treatment and outcome of female reproductive disorders, including tumoral and iatrogenic diseases.


Assuntos
Hormônio Liberador de Gonadotropina/fisiologia , Neurônios/fisiologia , Receptores LHRH/fisiologia , Reprodução/fisiologia , Feminino , Hormônio Liberador de Gonadotropina/análise , Gônadas/metabolismo , Humanos , Hipogonadismo , Hipotálamo , Doenças Ovarianas
3.
Theor Biol Med Model ; 9: 35, 2012 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-22870942

RESUMO

When hormones during the ovulatory cycle are shown in phase plane graphs, reported FSH and estrogen values form a specific pattern that resembles the leaning "&" symbol, while LH and progesterone (Pg) values form a "boomerang" shape. Graphs in this paper were made using data reported by Stricker et al. [Clin Chem Lab Med 2006;44:883-887]. These patterns were used to construct a simplistic model of the ovulatory cycle without the conventional "positive feedback" phenomenon. The model is based on few well-established relations:hypothalamic GnRH secretion is increased under estrogen exposure during two weeks that start before the ovulatory surge and lasts till lutheolysis.the pituitary GnRH receptors are so prone to downregulation through ligand binding that this must be important for their function.in several estrogen target tissue progesterone receptor (PgR) expression depends on previous estrogen binding to functional estrogen receptors (ER), while Pg binding to the expressed PgRs reduces both ER and PgR expression.Some key features of the presented model are here listed:High GnRH secretion induced by the recovered estrogen exposure starts in the late follicular phase and lasts till lutheolysis. The LH and FSH surges start due to combination of accumulated pituitary GnRH receptors and increased GnRH secretion. The surges quickly end due to partial downregulation of the pituitary GnRH receptors (64% reduction of the follicular phase pituitary GnRH receptors is needed to explain the reported LH drop after the surge). A strong increase in the lutheal Pg blood level, despite modest decline in LH levels, is explained as delayed expression of pituitary PgRs. Postponed pituitary PgRs expression enforces a negative feedback loop between Pg levels and LH secretions not before the mid lutheal phase.Lutheolysis is explained as a consequence of Pg binding to hypothalamic and pituitary PgRs that reduces local ER expression. When hypothalamic sensitivity to estrogen is diminished due to lack of local ERs, hypothalamus switches back to the low GnRH secretion rate, leading to low secretion of gonadotropins and to lutheolysis. During low GnRH secretion rates, previously downregulated pituitary GnRH receptors recover to normal levels and thus allow the next cycle.Possible implications of the presented model on several topics related to reproductive physiology are shortly discussed with some evolutionary aspects including the emergence of menopause.


Assuntos
Ciclo Menstrual/fisiologia , Modelos Biológicos , Ovulação/fisiologia , Ativinas/fisiologia , Evolução Biológica , Estrogênios/fisiologia , Retroalimentação Fisiológica , Feminino , Hormônio Foliculoestimulante/fisiologia , Humanos , Hipotálamo/fisiologia , Inibinas/fisiologia , Leptina/fisiologia , Hormônio Luteinizante/fisiologia , Luteólise/fisiologia , Masculino , Hipófise/fisiologia , Progesterona/fisiologia , Puberdade/fisiologia , Receptores de Estrogênio/fisiologia , Receptores LHRH/fisiologia , Receptores de Progesterona/fisiologia , Caracteres Sexuais
4.
Endocrinology ; 150(6): 2847-56, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19264870

RESUMO

In vertebrates, activation of the GnRH receptor is necessary to initiate the reproductive cascade. However, little is known about the characteristics of GnRH receptors before the vertebrates evolved. Recently genome sequencing was completed for amphioxus, Branchiostoma floridae. To understand the GnRH receptors (GnRHR) from this most basal chordate, which is also classified as an invertebrate, we cloned and characterized four GnRHR cDNAs encoded in the amphioxus genome. We found that incubation of GnRH1 (mammalian GnRH) and GnRH2 (chicken GnRH II) with COS7 cells heterologously expressing the amphioxus GnRHRs caused potent intracellular inositol phosphate turnover in two of the receptors. One of the two receptors displayed a clear preference for GnRH1 over GnRH2, a characteristic not previously seen outside the type I mammalian GnRHRs. Phylogenetic analysis grouped the four receptors into two paralogous pairs, with one pair grouping basally with the vertebrate GnRH receptors and the other grouping with the octopus GnRHR-like sequence and the related receptor for insect adipokinetic hormone. Pharmacological studies showed that octopus GnRH-like peptide and adipokinetic hormone induced potent inositol phosphate turnover in one of these other two amphioxus receptors. These data demonstrate the functional conservation of two distinct types of GnRH receptors at the base of chordates. We propose that one receptor type led to vertebrate GnRHRs, whereas the other type, related to the mollusk GnRHR-like receptor, was lost in the vertebrate lineage. This is the first report to suggest that distinct invertebrate and vertebrate GnRHRs are present simultaneously in a basal chordate, amphioxus.


Assuntos
Evolução Molecular , Invertebrados , Filogenia , Receptores LHRH/análise , Receptores LHRH/genética , Vertebrados , Sequência de Aminoácidos , Animais , Cordados , DNA Complementar/genética , Humanos , Dados de Sequência Molecular , Receptores LHRH/fisiologia , Reprodução/fisiologia , Transdução de Sinais/fisiologia
5.
Endocrinology ; 149(6): 3118-29, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18356273

RESUMO

GnRH and its structural variants bind to GnRH receptors from different species with different affinities and specificities. By investigating chimeric receptors that combine regions of mammalian and nonmammalian GnRH receptors, a greater understanding of how different domains influence ligand binding and receptor activation can be achieved. Using human-catfish and human-chicken chimeric receptors, we demonstrate the importance of extracellular loop conformation for ligand binding and agonist potency, providing further evidence for GnRH and GnRH II stabilization of distinct active receptor conformations. We demonstrate examples of GnRH receptor gain-of-function mutations that apparently improve agonist potency independently of affinity, implicating a role for extracellular loops in stabilizing the inactive receptor conformation. We also show that entire extracellular loop substitution can overcome the detrimental effects of localized mutations, thereby demonstrating the importance of considering the conformation of entire domains when drawing conclusions from point-mutation studies. Finally, we present evidence implicating the configuration of extracellular loops 2 and 3 in combination differentiating GnRH analog binding modes. Because there are two endogenous forms of GnRH ligand but only one functional form of full-length GnRH receptor in humans, understanding how GnRH and GnRH II can elicit distinct functional effects through the same receptor is likely to provide important insights into how these ligands can have differential effects in both physiological and pathological situations.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Receptores LHRH/química , Receptores LHRH/fisiologia , Sequência de Aminoácidos , Animais , Sítios de Ligação , Peixes-Gato , DNA Complementar/genética , Humanos , Fosfatos de Inositol/metabolismo , Cinética , Ligantes , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Especificidade da Espécie
6.
Endocrinology ; 149(6): 2899-906, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18325994

RESUMO

We demonstrated in a previous study that serum IL-8 concentrations were significantly higher in women with hot flashes than without hot flashes. To clarify the role of IL-8 in the pathoetiology of menopausal hot flashes, we examined the effect of rat cytokine-induced neutrophil chemoattractant (CINC), a member of the IL-8 family, on thermoregulation using ovariectomized (OVX) rats treated with intracerebroventricular (i.c.v.) injection of LHRH agonist (LHRHa) as a model of hot flashes. We found that: 1) expression of CINC mRNA was increased around the periventricular area in the hypothalamus at 1 h, and the serum CINC concentration was increased at 2 h after i.c.v. injection of LHRHa; 2) the increase in serum CINC concentration in hypophysectomized rats was significantly lower than that in sham-operated rats; 3) i.c.v. but not iv injection of CINC elevated the rectal temperature of OVX rats; 4) i.c.v. injection of LHRHa into OVX rats produced a rapid rise (maximal increase: 10-25 min) in tail skin temperature, and the elevation was augmented by injection of an anti-CINC antibody; and 5) changes in serum CINC concentration and skin temperature after i.c.v. injection of LHRHa were reversed by replacement of estradiol. In conclusion, the production of CINC in the hypothalamus due to LHRHa injection in OVX rats was increased after elevation of skin temperature, suggesting that CINC plays a key role in the homeostasis of body temperature. Disturbance of the thermoregulatory mechanism involving LHRH and CINC may be related to the pathoetiology of hot flashes.


Assuntos
Regulação da Temperatura Corporal , Quimiocina CXCL1/fisiologia , Quimiocinas CXC/fisiologia , Citocinas/fisiologia , Hormônio Liberador de Gonadotropina/fisiologia , Hipotálamo/fisiologia , Receptores LHRH/fisiologia , Animais , Feminino , Interleucina-8/sangue , Interleucina-8/fisiologia , Azeite de Oliva , Óleos de Plantas , Ratos , Ratos Sprague-Dawley
8.
Ann Urol (Paris) ; 39 Suppl 3: S37-45, 2005 Oct.
Artigo em Francês | MEDLINE | ID: mdl-16302709

RESUMO

Current data make it possible to relate idiopathic hypogonadotrophic hypogonadism to mutations affecting the GnRH I-1 receptor and also to new "loss-of-function" mutations concerning another receptor, GPR54. It now seems that mutations of the pituitary GnRH receptor are not the only explanation of most cases of sporadic isolated hypogonadotrophic hypogonadism, and, on the contrary, there are certain familial forms, where no mutation has been demonstrated, suggesting the potential involvement of other genes. The role of another G protein-coupled glycoprotein membrane receptor, GRP54, already known for its involvement as a metastasis suppressor, has been demonstrated. Bioclinical studies of families affected with the disorder by pheno/genotypic correlation demonstrated that "loss-of-function" mutations affecting the GPR54 gene coding for GRP54 are the cause of hypogonadotrophic hypogonadism. GPR54 therefore appears to be involved at hypothalamic and pituitary level. It is not involved in sexual differentiation, but may modulate GnRH secretion or affect its pituitary response. Further investigations are required to determine the levels of action of this receptor which may provide a new pharmacological target in the future.


Assuntos
Hormônio Liberador de Gonadotropina/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Receptores de Neuropeptídeos/genética , Membrana Celular/fisiologia , Proteínas de Ligação ao GTP/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Hipogonadismo/genética , Hipogonadismo/fisiopatologia , Hipotálamo/fisiologia , Hipófise/fisiologia , Receptores de Kisspeptina-1 , Receptores LHRH/fisiologia , Receptores de Neuropeptídeos/fisiologia , Diferenciação Sexual
9.
Ann Urol (Paris) ; 39 Suppl 3: S46-50, 2005 Oct.
Artigo em Francês | MEDLINE | ID: mdl-16302710

RESUMO

Although after the discovery of GnRH, research was initially directed towards the treatment of infertility, the development during the last twenty-five years of synthetic GnRH analogs has led to major advances in the diagnosis and treatment of endocrine disorders and cancer. Agonists are 50-100 times more potent than the natural neuropeptide and induce an intense and constant secretion of gonadotrophins, while their continuous administration induces hypophyseal desensitization with a fall in FSH and LH production leading to a reduction in sex hormones production and therefore chemical castration. This has been used for the treatment of precocious puberty, in vitro fertilization protocols and management of various hormone-dependent cancers such as prostate and breast cancer, a field where these indications are being continually extended. LHRH antagonists, used at doses higher than those of agonists, induce an immediate inhibition of gonadotrophin secretion and rapidly reduce sex steroid levels. Their development is more recent, and they have begun to find a role in prostatic diseases, cancer and benign prostatic hypertrophy. The discovery of GnRH receptors in various types of cancer has suggested a direct cytotoxic effect on cancer cells as well as the indirect hormonal effect. Research currently in the preclinical stage involves the use of combinations of ligand analogs and cytotoxic agents to increase the anti-tumoral specificity of chemotherapy and provide greater efficacy and reduced collateral toxicity. The management strategy of prostate, breast and ovarian cancers may therefore be considerably modified. Likewise, this concept of targeted chemotherapy using analogs acting as cytotoxic agent carriers up to the tumor site is the aim of research to evaluate somatostatin and bombesin.


Assuntos
Antineoplásicos/farmacologia , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/fisiologia , Receptores LHRH/fisiologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/fisiopatologia , Feminino , Humanos , Hipotálamo/fisiologia , Masculino , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/fisiopatologia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/fisiopatologia
10.
Anim Reprod Sci ; 88(1-2): 95-113, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15982835

RESUMO

Gonadotropin releasing hormone (GnRH) is a hypothalamic neuronal secretory decapeptide that plays a pivotal role in mammalian reproduction. GnRH and its analogues are used extensively in the treatment of hormone dependent diseases and assisted reproductive technology. Fourteen structural variants and three different forms of GnRH, named as hypothalamic GnRH or GnRH-I, mid brain GnRH or GnRH-II and GnRH-III across various species of protochordates and vertebrates have been recognised. The hormone acts by binding to cell surface transmembrane G protein coupled receptors (GPCRs) and activates Gq/11 subfamily of G proteins. Although hypothalamus and pituitary are the principal source and target sites for GnRH, several reports have recently suggested extra-hypothalamic GnRH and GnRH receptors in various reproductive tissues such as ovaries, placenta, endometrium, oviducts, testes, prostrate, and mammary glands. GnRH-II appears to be predominantly expressed in extra pituitary reproductive tissues where it produces its effect by PLC, PKA2, PLD, and AC cell signalling pathways. In these tissues, GnRH is considered to act by autocrine or paracrine manner and regulate ovarian steroidogenesis by having stimulatory as well as inhibitory effect on the production of steroid hormones and apoptosis in ovarian follicle and corpus luteum. In male gonads, GnRH has been shown to cause a direct stimulatory effect on basal steroidogenesis and an inhibitory effect on gonadotropin-stimulated androgen biosynthesis. Recent studies have shown that GnRH is more abundantly present in ovarian, endometrial and prostrate carcinomas. The presence of type-II GnRH receptors in reproductive tissues (e.g. gonads, prostrate, endometrium, oviduct, placenta, and mammary glands) suggests existence of distinct role(s) for type-II GnRH molecule in these tissues. The existence of different GnRH forms indicates the presence of distinctive cognate receptors types in vertebrates and is a productive area of research and may contribute to the development of new generation of GnRH analogues with highly selective and controlled action on different reproductive tissues and the target-specific GnRH analogues could be developed.


Assuntos
Hormônio Liberador de Gonadotropina/análise , Hormônio Liberador de Gonadotropina/fisiologia , Reprodução , Animais , Apoptose , Feminino , Expressão Gênica , Hormônio Liberador de Gonadotropina/genética , Humanos , Hipotálamo/química , Masculino , Especificidade de Órgãos , Ovário/química , Ovário/citologia , Ovário/efeitos dos fármacos , Hipófise/química , Hipófise/fisiologia , Placenta/química , Gravidez , Receptores LHRH/análise , Receptores LHRH/genética , Receptores LHRH/fisiologia , Testículo/química
11.
J Soc Biol ; 198(1): 80-7, 2004.
Artigo em Francês | MEDLINE | ID: mdl-15146960

RESUMO

The acquisition of a sexually dimorphic phenotype is a critical event in mammalian development. Hypogonadotropic hypogonadism (HH) results from impaired secretion of GnRH. The patients display with delayed puberty, micropenis and cryptorchidism in the male reflecting gonadotropin insufficiency, and amenorrhea in the female. Kallmann's syndrome (KS) is defined by the association of HH and anosmia or hyposmia (absent smelling sense). Segregation analysis in familial cases has demonstrated diverse inheritance patterns, suggesting the existence of several genes regulating GnRH secretion. The X-linked form of the disease was associated with a genetic defect in the KALI gene located on the Xp22.3 region. KAL1 gene encodes an extracellular matrix glycoprotein anosmin-1, which facilitates neuronal growth and migration. Abnormalities in the migratory processes of the GnRH neurons with the olfactory neurons explain the association of HH with anosmia. Recently, mutations in the FGF recepteur 1 (FGFR1) gene were found in KS with autosomal dominant mode of inheritance. The role of FGFR1 in the function of reproduction requires further investigation. Besides HH with anosmia, there are isolated HH (IHH). No human GnRH mutations have been reported although hypogonadal mice due to a GnRH gene deletion exist. In patients with idiopathic HH and without anosmia an increasing number of GnRH receptor (GnRHR) mutations have been described which represent about 50% of familial cases. The clinical features are highly variable and there is a good relationship between genotype and phenotype. A complete loss of function is associated with the most severe phenotype with resistance to pulsatile GnRH treatment, absence of puberty and cryptorchidism in the male. In contrast, milder loss of function mutations causes incomplete failure of pubertal development. The preponderant role of GnRH in the secretion of LH by the gonadotrophs explains the difference of the phenotype between male and female with partial GnRH resistance. Affected females can have spontaneous telarche and normal breast development while affected males exhibit no pubertal development but normal testis volume, a feature described as "fertile-eunuch". High-dose pulsatile GnRH has been used to induce ovulation. Another gene, called GPR54, responsible for idiopathic HH has been recently described by segregation analysis in two different consanguineous families. The GPR54 gene is an orphan receptor, and its putative ligand is the product of the KISS-1 gene, called metastine. Their roles in the function of reproduction are still unknown.


Assuntos
Hormônio Liberador de Gonadotropina/deficiência , Hipogonadismo/genética , Receptores LHRH/deficiência , Sequência de Aminoácidos , Animais , Movimento Celular , Consanguinidade , Proteínas da Matriz Extracelular/deficiência , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/fisiologia , Feminino , Heterogeneidade Genética , Genótipo , Hormônio Liberador de Gonadotropina/fisiologia , Humanos , Hipogonadismo/fisiopatologia , Hipotálamo/citologia , Hipotálamo/embriologia , Síndrome de Kallmann/genética , Síndrome de Kallmann/fisiopatologia , Kisspeptinas , Masculino , Camundongos , Modelos Biológicos , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Transtornos do Olfato/complicações , Transtornos do Olfato/genética , Fenótipo , Mutação Puntual , Proteínas/fisiologia , Receptores Proteína Tirosina Quinases/deficiência , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/fisiologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Receptores de Fatores de Crescimento de Fibroblastos/deficiência , Receptores de Fatores de Crescimento de Fibroblastos/genética , Receptores de Fatores de Crescimento de Fibroblastos/fisiologia , Receptores Acoplados a Proteínas G , Receptores de Kisspeptina-1 , Receptores LHRH/química , Receptores LHRH/genética , Receptores LHRH/fisiologia , Receptores de Neuropeptídeos/deficiência , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/fisiologia , Proteínas Supressoras de Tumor
12.
J Gynecol Obstet Biol Reprod (Paris) ; 33(6 Pt 2): 3S7-10, 2004 Oct.
Artigo em Francês | MEDLINE | ID: mdl-15643677

RESUMO

GnRH is the native decapeptide which initiates the reproductive cascade. It is synthesized in a loose network of hypothalamic neurons and released into the hypothalamo-pituitary portal blood system in a pulsatile manner. The main physiologic actions of GnRH include the synthesis and release of LH and FSH. Analogs are synthetic versions of GnRH with various amino acid substitutions. These substitutions serve to increase their half-life and to increase their affinity for the GnRH receptor. There are two types of analog: GnRH agonists and GnRH antagonists. GnRH agonists behave like GnRH and are initially stimulatory ("flare up"). GnRH antagonists block the effects of GnRH and are inhibitory. When GnRH antagonists bind to the GnRH receptor they do not initiate the normal cascade of intracellular events, they prevent GnRH from gaining access to the receptor and prevent the above cascade from occuring. Consequently there is no "Flare Effect" and levels of LH and FSH begin immediately to fall. GnRH antagonists do not cause GnRH receptor downregulation: the pituitary remains responsive to GnRH or GnRH agonist administration. The degree of suppression of circulating LH and FSH is dependent on circulating levels of the GnRH antagonist. Administration of GnRH antagonist produces suppression of endogenous LH and FSH at all phases of the cycle. The degree of suppression is dependent on the amount of GnRH antagonist administered. The suppression of endogenous LH and FSH produced by GnRH antagonist can be overridden by GnRH or GnRH agonist.


Assuntos
Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Ciclo Menstrual/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Feminino , Hormônio Foliculoestimulante/metabolismo , Hormônio Liberador de Gonadotropina/agonistas , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/química , Hormônio Liberador de Gonadotropina/fisiologia , Humanos , Hipotálamo/fisiologia , Hormônio Luteinizante/metabolismo , Ciclo Menstrual/fisiologia , Receptores LHRH/antagonistas & inibidores , Receptores LHRH/efeitos dos fármacos , Receptores LHRH/fisiologia
13.
J Biol Chem ; 279(1): 414-20, 2004 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-14573593

RESUMO

Numerous external stimuli, including G protein-coupled receptor agonists, cytokines, growth factors, and steroids activate mitogen-activated protein kinases (MAPKs) through phosphorylation of the epidermal growth factor receptor (EGF-R). In immortalized hypothalamic neurons (GT1-7 cells), agonist binding to the gonadotropin-releasing hormone receptor (GnRH-R) causes phosphorylation of MAPKs that is mediated by protein kinase C (PKC)-dependent transactivation of the EGF-R. An analysis of the mechanisms involved in this process showed that GnRH stimulation of GT1-7 cells causes release/shedding of the soluble ligand, heparin binding epidermal growth factor (HB-EGF), as a consequence of metalloprotease activation. GnRH-induced phosphorylation of the EGF-R and, subsequently, of Shc, ERK1/2, and its dependent protein, p90RSK-1 (p90 ribosomal S6 kinase 1 or RSK-1), was abolished by metalloprotease inhibition. Similarly, blockade of the effect of HB-EGF with the selective inhibitor CRM197 or a neutralizing antibody attenuated signals generated by GnRH and phorbol 12-myristate 13-acetate, but not those stimulated by EGF. In contrast, phosphorylation of the EGF-R, Shc, and ERK1/2 by EGF and HB-EGF was independent of PKC and metalloprotease activity. The signaling characteristics of HB-EGF closely resembled those of GnRH and EGF in terms of the phosphorylation of EGF-R, Shc, ERK1/2, and RSK-1 as well as the nuclear translocation of RSK-1. However, neither the selective Src kinase inhibitor PP2 nor the overexpression of negative regulatory Src kinase and dominant negative Pyk2 had any effect on HB-EGF-induced responses. In contrast to GT1-7 cells, human embryonic kidney 293 cells expressing the GnRH-R did not exhibit metalloprotease induction and EGF-R transactivation during GnRH stimulation. These data indicate that the GnRH-induced transactivation of the EGF-R and the subsequent ERK1/2 phosphorylation result from ectodomain shedding of HBEGF through PKC-dependent activation of metalloprotease(s) in neuronal GT1-7 cells.


Assuntos
Receptores ErbB/genética , Metaloproteases/metabolismo , Neurônios/fisiologia , Animais , Linhagem Celular , Clonagem Molecular , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/farmacologia , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Humanos , Hipotálamo/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Receptores de Superfície Celular/metabolismo , Receptores LHRH/fisiologia , Proteínas Recombinantes/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , Ativação Transcricional , Transfecção
14.
J Biol Chem ; 278(21): 19118-26, 2003 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-12642580

RESUMO

The duration as well as the magnitude of mitogen-activated protein kinase activation has been proposed to regulate gene expression and other specific intracellular responses in individual cell types. Activation of ERK1/2 by the hypothalamic neuropeptide gonadotropin-releasing hormone (GnRH) is relatively sustained in alpha T3-1 pituitary gonadotropes and HEK293 cells but is transient in immortalized GT1-7 neurons. Each of these cell types expresses the epidermal growth factor receptor (EGFR) and responds to EGF stimulation with significant but transient ERK1/2 phosphorylation. However, GnRH-induced ERK1/2 phosphorylation caused by EGFR transactivation was confined to GT1-7 cells and was attenuated by EGFR kinase inhibition. Neither EGF nor GnRH receptor activation caused translocation of phospho-ERK1/2 into the nucleus in GT1-7 cells. In contrast, agonist stimulation of GnRH receptors expressed in HEK293 cells caused sustained phosphorylation and nuclear translocation of ERK1/2 by a protein kinase C-dependent but EGFR-independent pathway. GnRH-induced activation of ERK1/2 was attenuated by the selective Src kinase inhibitor PP2 and the negative regulatory C-terminal Src kinase in GT1-7 cells but not in HEK293 cells. In GT1-7 cells, GnRH stimulated phosphorylation and nuclear translocation of the ERK1/2-dependent protein, p90RSK-1 (RSK-1). These results indicate that the duration of ERK1/2 activation depends on the signaling pathways utilized by GnRH in specific target cells. Whereas activation of the Gq/protein kinase C pathway in HEK293 cells causes sustained phosphorylation and translocation of ERK1/2 to the nucleus, transactivation of the EGFR by GnRH in GT1-7 cells elicits transient ERK1/2 signals without nuclear accumulation. These findings suggest that transactivation of the tightly regulated EGFR can account for the transient ERK1/2 responses that are elicited by stimulation of certain G protein-coupled receptors.


Assuntos
Receptores ErbB/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Receptores LHRH/fisiologia , Ativação Transcricional , Quinases da Família src/genética , Animais , Transporte Biológico , Linhagem Celular , Linhagem Celular Transformada , Núcleo Celular/metabolismo , Ativação Enzimática/efeitos dos fármacos , Expressão Gênica , Genes fos/genética , Hormônio Liberador de Gonadotropina/farmacologia , Hipotálamo , Immunoblotting , Imuno-Histoquímica , Fosfatos de Inositol/análise , Camundongos , Proteína Quinase 3 Ativada por Mitógeno , Neurônios , Fosforilação , Proteína Quinase C/metabolismo , Receptores LHRH/efeitos dos fármacos , Receptores LHRH/genética , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Transdução de Sinais , Acetato de Tetradecanoilforbol/administração & dosagem , Transfecção
15.
Trends Endocrinol Metab ; 14(1): 35-43, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12475610

RESUMO

Hypothalamic gonadotrophin-releasing hormone (GnRH I), which is of a variable structure in vertebrates, is the central regulator of the reproductive system through its stimulation of gonadotrophin release from the pituitary. A second form of GnRH (GnRH II) is ubiquitous and conserved in structure from fish to humans, suggesting that it has important functions and a discriminating receptor that selects against structural change. GnRH II is distributed in discrete regions of the central and peripheral nervous systems and in nonneural tissues. The cognate receptor for GnRH II has recently been cloned from amphibians and mammals. It is highly selective for GnRH II, has a similar distribution to GnRH II in the nervous system and, notably, in areas associated with sexual behaviour. It is also found in reproductive tissues. An established function of GnRH II is in the inhibition of M currents (K(+) channels) through the GnRH II receptor in the amphibian sympathetic ganglion, and it might act through this mechanism as a neuromodulator in the central nervous system. The conservation of structure over 500 million years and the wide tissue distribution of GnRH II suggest that it has a variety of reproductive and nonreproductive functions and will be a productive area of research.


Assuntos
Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/metabolismo , Sistema Nervoso/química , Receptores LHRH/metabolismo , Sequência de Aminoácidos , Animais , Hormônio Liberador de Gonadotropina/química , Hormônio Liberador de Gonadotropina/fisiologia , Humanos , Hipotálamo/química , Dados de Sequência Molecular , Sistema Nervoso/metabolismo , Receptores LHRH/fisiologia , Transdução de Sinais , Distribuição Tecidual
16.
Front Neuroendocrinol ; 24(4): 279-95, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14726258

RESUMO

It is now well known that different forms of GnRH coexist in the same vertebrate species. In humans, two forms of GnRH have been identified so far. The first form corresponds to the hypophysiotropic decapeptide, and is now called GnRH-I. The second form has been initially identified in the chicken brain, and it is referred to as GnRH-II. GnRH-I binds to and activates specific receptors, belonging to the 7 transmembrane (7TM) domain superfamily, present on pituitary gonadotropes. These receptors (type I GnRH receptors) are coupled to the Gq/11/PLC intracellular signalling pathway. A receptor specific for GnRH-II (type II GnRH receptor) has been identified in non-mammalian vertebrates as well as in primates, but not yet in humans. In the last 10-15 years experimental evidence has been accumulated indicating that GnRH-I is expressed, together with its receptors, in tumors of the reproductive tract (prostate, breast, ovary, and endometrium). In these hormone-related tumors, activation of type I GnRH receptors consistently decreases cell proliferation, mainly by interfering with the mitogenic activity of stimulatory growth factors (e.g., EGF, IGF). Recent data seem to suggest that GnRH-I might also reduce the migratory and invasive capacity of cancer cells, possibly by affecting the expression and/or activity of cell adhesion molecules and of enzymes involved in the remodelling of the extracellular matrix. These observations point to GnRH-I as an autocrine negative regulatory factor on tumor growth progression and metastatization. Extensive research has been performed to clarify the molecular mechanisms underlying the peculiar antitumor activity of GnRH-I. Type I GnRH receptors in hormone-related tumors correspond to those present at the pituitary level in terms of cDNA nucleotide sequence and protein molecular weight, but do not share the same pharmacological profile in terms of binding affinity for the different synthetic GnRH-I analogs. Moreover, the classical intracellular signalling pathway mediating the stimulatory activity of the decapeptide on gonadotropin synthesis and secretion is not involved in its inhibitory activity on hormone-related tumor growth. In these tumors, type I GnRH receptors are coupled to the Gi-cAMP, rather than the Gq/11-PLC, signal transduction pathway. Recently, we have reported that GnRH-I and type I GnRH receptors are expressed also in tumors not related to the reproductive system, such as melanoma. Also in melanoma cells, GnRH-I behaves as a negative regulator of tumor growth and progression. Interestingly, the biochemical and pharmacological profiles of type I GnRH receptors in melanoma seem to correspond to those of the receptors at pituitary level. The data so far reported on the expression and on the possible functions of GnRH-II in humans are still scanty. The decapeptide has been identified, together with a 'putative' type II GnRH receptor, both in the central nervous system and in peripheral structures, such as tissues of the reproductive tract (both normal and tumoral). The specific biological functions of GnRH-II in humans are presently under investigation.


Assuntos
Regulação Neoplásica da Expressão Gênica/fisiologia , Hormônio Liberador de Gonadotropina/fisiologia , Receptores LHRH/fisiologia , Neoplasias Urogenitais/genética , Neoplasias Urogenitais/metabolismo , Animais , Progressão da Doença , Hormônio Liberador de Gonadotropina/análogos & derivados , Humanos , Sistema Hipotálamo-Hipofisário/metabolismo , Hipotálamo/metabolismo , Hipófise/metabolismo
17.
Endocrinology ; 142(5): 1737-43, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11316736

RESUMO

GnRH acts via GnRH receptors (GnRH-R) in the pituitary to cause the release of gonadotropins that regulate vertebrate reproduction. In the teleost fish, Haplochromis burtoni, reproduction is socially regulated through the hypothalamus-pituitary-gonadal axis, making the pituitary GnRH-R a likely site of action for this control. As a first step toward understanding the role of GnRH-R in the social control of reproduction, we cloned and sequenced candidate GnRH-R complementary DNAs from H. burtoni tissue. We isolated a complementary DNA that predicts a peptide encoding a G protein-coupled receptor that shows highest overall identity to other fish type I GnRH-R (goldfish IA and IB and African catfish). Functional testing of the expressed protein in vitro confirmed high affinity binding of multiple forms of GNRH: Localization of GnRH-R messenger RNA using RT-PCR revealed that it is widely distributed in the brain and retina as well as elsewhere in the body. Taken together, these data suggest that this H. burtoni GnRH receptor probably interacts in vivo with all three forms of GNRH:


Assuntos
Peixes/fisiologia , Receptores LHRH/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , DNA Complementar/química , Fosfatos de Inositol/metabolismo , Masculino , Dados de Sequência Molecular , Filogenia , RNA Mensageiro/análise , Receptores LHRH/análise , Receptores LHRH/química
18.
J Biol Chem ; 276(16): 12736-43, 2001 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-11278883

RESUMO

The ability of G-protein-coupled receptors (GPCRs) to interact to form new functional structures, either forming oligomers with themselves or forming associations with other intracellular proteins, has important implications for the regulation of cellular events; however, little is known about how this occurs. Here, we have employed a newly emerging technology, bioluminescence resonance energy transfer (BRET), used to study protein-protein interactions in living cells, to demonstrate that the thyrotropin-releasing hormone receptor (TRHR) forms constitutive homo-oligomers. This formation of TRHR homo-oligomers in the absence of ligand was shown by demonstration of an energy transfer between TRHR molecules fused to either donor, Renilla luciferase (Rluc) or acceptor, enhanced yellow fluorescent protein (EYFP) molecules. This interaction was shown to be specific, since energy transfer was not detected between co-expressed tagged TRHRs and either complementary tagged gonadotropin-releasing hormone (GnRH) or beta(2)-adrenergic receptors. Furthermore, generation of a BRET signal between the TRHRs could only be inhibited by co-expression of the wild-type TRHR and not by other GPCRs. Agonist stimulation led to a time- and dose-dependent increase in the amount of energy transfer. Inhibition of receptor internalization by co-expression of dynamin mutant K44A did not affect the interaction between TRHRs, suggesting that clustering of receptors within clathrin-coated pits is not sufficient for energy transfer to occur. BRET also provided evidence for the agonist-induced oligomerization of another GPCR, the GnRH receptor (GnRHR), and the presence of an agonist-induced interaction of the adaptor protein, beta-arrestin, with TRHR and the absence of an interaction of beta-arrestin with GnRHR. This study supports the usefulness of BRET as a powerful tool for studying GPCR aggregations and receptor/protein interactions in general and presents evidence that the functioning unit of TRHRs exists as homomeric complexes.


Assuntos
Receptores do Hormônio Liberador da Tireotropina/química , Receptores do Hormônio Liberador da Tireotropina/fisiologia , Pamoato de Triptorrelina/análogos & derivados , Substituição de Aminoácidos , Animais , Arrestinas/metabolismo , Proteínas de Bactérias/análise , Proteínas de Bactérias/genética , Células COS , Linhagem Celular , Membrana Celular/fisiologia , Chlorocebus aethiops , Invaginações Revestidas da Membrana Celular/fisiologia , Transferência de Energia , Humanos , Radioisótopos do Iodo , Luciferases/análise , Luciferases/genética , Medições Luminescentes , Proteínas Luminescentes/análise , Proteínas Luminescentes/genética , Substâncias Macromoleculares , Mutagênese Sítio-Dirigida , Ratos , Receptores LHRH/agonistas , Receptores LHRH/química , Receptores LHRH/fisiologia , Receptores do Hormônio Liberador da Tireotropina/agonistas , Proteínas Recombinantes de Fusão/agonistas , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Transfecção , Pamoato de Triptorrelina/farmacocinética , beta-Arrestinas
19.
Indian J Physiol Pharmacol ; 45(4): 395-407, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11883145

RESUMO

Gonadotropin-releasing hormone (GnRH) secretion from the hypothalamus is pivotal to the regulation of reproductive physiology in vertebrates. The characteristic periodic secretion of gonadotropin releasing hormone (GnRH) from the medial basal hypothalamus (MBH), at the rate of one pulse an hour is essential for the maintenance of the menstrual cycle. These pulses are due to oscillations in the electrical activity of the GnRH pulse generator in the MBH. The GnRH pulse generator is under the influence of an assortment of interactions of multiple neural, hormonal and environmental inputs to the hypothalamus. Hence, a number of conditions such as stress, drug intake, exercise, sleep affect the activity of this pulse generator. Any deviation of normal frequency results in disruption of normal cycle. The cycle can become anovulatory in the hypothalamic lesions and can be restored by exogenous administration of pulsatile GnRH. Of late, studies have shown that pulse generator activity is also maintained by specific metabolic signals meant for energy homeostasis. Studies are in progress to work out cellular basis of GnRH pulse generator's rhythmic activation and role of Ca++ as second messenger for GnRH stimulated gonadotropin release. New concepts are emerging to find the existence of an FSH releasing factor, which independently regulates the activity of FSH.


Assuntos
Sistema Hipotálamo-Hipofisário/fisiologia , Ciclo Menstrual , Ovário/fisiologia , Monoaminas Biogênicas/fisiologia , Feminino , Hormônio Liberador de Gonadotropina/fisiologia , Humanos , Hipotálamo/embriologia , Masculino , Hipófise/embriologia , Receptores LHRH/fisiologia
20.
Endocrinology ; 139(4): 1781-8, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9528962

RESUMO

GnRH stimulates gonadotropin secretion, which desensitizes unless the releasing hormone is secreted or administered in a pulsatile fashion. The mechanism of desensitization is unknown, but as the GnRH receptor is G protein coupled, it might involve G protein-coupled receptor kinases (GRKs). Such kinases phosphorylate the intracellular regions of seven-transmembrane receptors, permitting beta-arrestin to bind, which prevents the receptor from activating G proteins. Here, we tested the effect of GRKs and beta-arrestins on GnRH-induced inositol trisphosphate (IP3) production in COS cells transfected with the GnRH receptor complementary DNA. GRK2, -3, and -6 overexpression inhibited IP3 production by 50-75% during the 30 sec of GnRH treatment. Coexpression of GRK2 and beta-arrestin-2 suppressed GnRH-induced IP3 production more than that of either alone. Immunocytochemical staining of rat anterior pituitary revealed that all cells expressed GRK2, -3, and -6; all cells also expressed the beta-arrestins. Western blots on cytosolic extracts of rat pituitaries revealed the presence of GRK2/3 and beta-arrestin-1 and -2. The expression of GRKs and beta-arrestins by gonadotropes and their inhibition of GnRH-stimulated IP3 production in COS-1 cells expressing the GnRH receptor suggest a potential regulatory role for the GRK/beta arrestin paradigm in GnRH receptor signaling.


Assuntos
Arrestinas/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptores LHRH/fisiologia , Transdução de Sinais , Animais , Arrestinas/análise , Células COS , Bovinos , Hormônio Liberador de Gonadotropina/farmacologia , Imuno-Histoquímica , Fosfatos de Inositol/biossíntese , Cinética , Fosforilação , Adeno-Hipófise/química , Ratos , Receptores Proteína Tirosina Quinases/análise , Transfecção , beta-Arrestina 1 , beta-Arrestina 2 , beta-Arrestinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA