Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 142
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
BMC Cancer ; 22(1): 133, 2022 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-35109816

RESUMEN

BACKGROUND: Gonadotropin-releasing hormone (GnRH) receptor, a rhodopsin-like G-protein coupled receptor (GPCR) family member involved in GnRH signaling, is reported to be expressed in several tumors including glioblastoma multiforme (GBM), one of the most malignant and aggressive forms of primary brain tumors. However, the molecular targets associated with GnRH receptor are not well studied in GBM or in other cancers. The present study aims at investigating the effect of GnRH agonist (Gosarelin acetate) on cell proliferation and associated signaling pathways in GBM cell line, LN229. METHODS: LN229 cells were treated with different concentrations of GnRH agonist (10-10 M to 10-5 M) and the effect on cell proliferation was analyzed by cell count method. Further, total protein was extracted from control and GnRH agonist treated cells (with maximum reduction in cell proliferation) followed by trypsin digestion, labeling with iTRAQ reagents and LC-MS/MS analysis to identify differentially expressed proteins. Bioinformatic analysis was performed for annotation of proteins for the associated molecular function, altered pathways and network analysis using STRING database. RESULTS: The treatment with different concentrations of GnRH agonist showed a reduction in cell proliferation with a maximum reduction of 48.2% observed at 10-6 M. Quantitative proteomic analysis after GnRH agonist treatment (10-6 M) led to the identification of a total of 29 differentially expressed proteins with 1.3-fold change (23 upregulated, such as, kininogen-1 (KNG1), alpha-2-HS-glycoprotein (AHSG), alpha-fetoprotein (AFP), and 6 downregulated, such as integrator complex subunit 11 (CPSF3L), protein FRG1 (FRG1). Some of them are known [KNG1, AHSG, AFP] while others such as inter-alpha-trypsin inhibitor heavy chain H2 (ITIH2), ITIH4, and LIM domain-containing protein 1 (LIMD1) are novel to GnRH signaling pathway. Protein-protein interaction analysis showed a direct interaction of KNG1, a hub molecule, with GnRH, GnRH receptor, EGFR and other interactors including ITIH2, ITIH4 and AHSG. Overexpression of KNG1 after GnRH agonist treatment was validated using Western blot analysis, while a significant inhibition of EGFR was observed after GnRH agonist treatment. CONCLUSIONS: The study suggests a possible link of GnRH signaling with EGFR signaling pathways likely via KNG1. KNG1 inhibitors may be investigated independently or in combination with GnRH agonist for therapeutic applications.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Proliferación Celular/efectos de los fármacos , Glioblastoma/metabolismo , Hormona Liberadora de Gonadotropina/biosíntesis , Receptores LHRH/biosíntesis , Animales , Antineoplásicos Hormonales/farmacología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Cromatografía Liquida , Biología Computacional , Glioblastoma/genética , Glioblastoma/patología , Hormona Liberadora de Gonadotropina/agonistas , Hormona Liberadora de Gonadotropina/genética , Goserelina/farmacología , Humanos , Proteómica/métodos , Receptores LHRH/genética , Transducción de Señal/efectos de los fármacos , Espectrometría de Masas en Tándem
2.
Annu Rev Neurosci ; 35: 133-51, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22524786

RESUMEN

In the course of evolution, social behavior has been a strikingly potent selective force in shaping brains to control action. Physiological, cellular, and molecular processes reflect this evolutionary force, particularly in the regulation of reproductive behavior and its neural circuitry. Typically, experimental analysis is directed at how the brain controls behavior, but the brain is also changed by behavior over evolution, during development, and through its ongoing function. Understanding how the brain is influenced by behavior offers unusual experimental challenges. General principles governing the social regulation of the brain are most evident in the control of reproductive behavior. This is most likely because reproduction is arguably the most important event in an animal's life and has been a powerful and essential selective force over evolution. Here I describe the mechanisms through which behavior changes the brain in the service of reproduction using a teleost fish model system.


Asunto(s)
Encéfalo/fisiología , Conducta Sexual Animal/fisiología , Conducta Social , Controles Informales de la Sociedad , Animales , Cíclidos , Regulación de la Expresión Génica/fisiología , Hormonas Esteroides Gonadales/biosíntesis , Gonadotropinas/biosíntesis , Modelos Biológicos , Receptores LHRH/biosíntesis , Espermatogénesis/genética
3.
Neurourol Urodyn ; 37(5): 1574-1582, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-30133853

RESUMEN

AIM: To evaluate the effects of a treatment with leuprolide acetate (LA) on bladder overactivity as well as the expression of gonadotropin releasing hormone receptor (GnRH-R), and neurofilaments NF68 and NF200 in female rats with overactive bladder induced by castration. METHODS: Changes in the urodynamic parameters were determined in SHAM, ovariectomized (OVX) and ovariectomized rats treated with LA (OVX-LA). A semi-quantitative analysis for the expression pattern of GnRH-R and neurofilaments NF68 and NF200 were determined. RESULTS: Forty-three days after ovariectomy, rats from the OVX group have significant lower values for intercontractile interval (ICI) and compliance (C); as well as higher values for basal bladder pressure (BP) and frequency of non-voiding contractions (NVC). The systemic application of LA increased voiding volume (Vv) and pressure threshold (ThP) in the OVX-LA animals. The application of LA reduced the high frequency of NVC in the OVX rats. No significant differences were found for Vv and NVCs between the OVX-LA vs SHAM groups. At the mid part of the bladder, the presence of GnRH-R was evidenced in the urothelium of the SHAM group. The OVX animals showed different pattern of immunolabeling for GnRH-R as well as for neurofilaments NF200 and NF68, whereas in the OVX-LA group the immunofluorescence pattern was similar to the one seen in SHAM bladders (P < 0.05 for OVX vs OVX + LA). CONCLUSIONS: the results suggest that systemic application of LA can improve bladder dysfunction in castrated rats, and perhaps considered as a treatment for overactive bladder conditions secondary to menopause.


Asunto(s)
Leuprolida/farmacología , Ovariectomía , Receptores LHRH/agonistas , Urodinámica/efectos de los fármacos , Animales , Adaptabilidad/efectos de los fármacos , Femenino , Contracción Muscular/efectos de los fármacos , Proteínas de Neurofilamentos/biosíntesis , Proteínas de Neurofilamentos/genética , Ratas , Ratas Wistar , Receptores LHRH/biosíntesis , Vejiga Urinaria/efectos de los fármacos , Vejiga Urinaria/metabolismo , Urotelio/efectos de los fármacos , Urotelio/metabolismo
4.
Reprod Fertil Dev ; 30(4): 672-680, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29019791

RESUMEN

This study aimed to explain how prolonged inhibition of central dopaminergic activity affects the cellular processes governing gonadotrophin-releasing hormone (GnRH) and LH secretion in anoestrous sheep. For this purpose, the study included two experimental approaches: first, we investigated the effect of infusion of sulpiride, a dopaminergic D2 receptor antagonist (D2R), on GnRH and GnRH receptor (GnRHR) biosynthesis in the hypothalamus and on GnRHR in the anterior pituitary using an immunoassay. This analysis was supplemented by analysis of plasma LH levels by radioimmunoassay. Second, we used real-time polymerase chain reaction to analyse the influence of sulpiride on the levels of kisspeptin (Kiss1) mRNA in the preoptic area and ventromedial hypothalamus including arcuate nucleus (VMH/ARC), and RFamide-related peptide-3 (RFRP-3) mRNA in the paraventricular nucleus (PVN) and dorsomedial hypothalamic nucleus. Sulpiride significantly increased plasma LH concentration and the levels of GnRH and GnRHR in the hypothalamic-pituitary unit. The abolition of dopaminergic activity resulted in a significant increase in transcript level of Kiss1 in VMH/ARC and a decrease of RFRP-3 in PVN. The study demonstrates that dopaminergic neurotransmission through D2R is involved in the regulatory pathways of GnRH and GnRHR biosynthesis in the hypothalamic-pituitary unit of anoestrous sheep, conceivably via mechanisms in which Kiss1 and RFRP-3 participate.


Asunto(s)
Anestro/metabolismo , Antagonistas de los Receptores de Dopamina D2/farmacología , Hormona Liberadora de Gonadotropina/biosíntesis , Kisspeptinas/metabolismo , Neuropéptidos/metabolismo , Receptores LHRH/biosíntesis , Sulpirida/farmacología , Animales , Femenino , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Hormona Luteinizante/sangre , Adenohipófisis/efectos de los fármacos , Adenohipófisis/metabolismo , Ovinos
5.
Transgenic Res ; 26(4): 567-575, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28534229

RESUMEN

Swine are the only livestock species that produce both the second mammalian isoform of gonadotropin-releasing hormone (GNRH2) and its receptor (GNRHR2). Previously, we reported that GNRH2 and GNRHR2 mediate LH-independent testosterone secretion from porcine testes. To further explore this ligand-receptor complex, a pig model with reduced GNRHR2 expression was developed. Small hairpin RNA sequences targeting porcine GNRHR2 were subcloned into a lentiviral-based vector, lentiviral particles were generated and microinjected into the perivitelline space of zygotes, and embryos were transferred into a recipient. One GNRHR2 knockdown (KD) female was born that subsequently produced 80 piglets from 6 litters with 46 hemizygous progeny (57% transgenic). Hemizygous GNRHR2 KD (n = 10) and littermate control (n = 7) males were monitored at 40, 100, 150, 190, 225 and 300 days of age; body weight and testis size were measured and serum was isolated and assayed for testosterone and luteinizing hormone (LH) concentrations. Body weight of GNRHR2 KD boars was not different from littermate controls (P = 0.14), but testes were smaller (P < 0.05; 331.8 vs. 374.8 cm3, respectively). Testosterone concentrations tended (P = 0.06) to be reduced in GNRHR2 KD (1.6 ng/ml) compared to littermate control (4.2 ng/ml) males, but LH levels were similar (P = 0.47). The abundance of GNRHR2 mRNA was reduced (P < 0.001) by 69% in testicular tissue from mature GNRHR2 KD (n = 5) versus littermate control (n = 4) animals. These swine represent the first genetically-engineered model to elucidate the function of GNRH2 and its receptor in mammals.


Asunto(s)
Animales Modificados Genéticamente/genética , Hormona Liberadora de Gonadotropina/genética , Receptores LHRH/genética , Animales , Animales Modificados Genéticamente/crecimiento & desarrollo , Femenino , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Hormona Liberadora de Gonadotropina/biosíntesis , Hemicigoto , Hormona Luteinizante/sangre , Masculino , ARN Interferente Pequeño/genética , Receptores LHRH/biosíntesis , Porcinos/genética , Porcinos/crecimiento & desarrollo , Testículo/crecimiento & desarrollo , Testículo/metabolismo , Testosterona/sangre
6.
Can J Physiol Pharmacol ; 95(2): 178-184, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27901351

RESUMEN

This study was performed to explain how the molecular processes governing the biosynthesis of gonadotropin-releasing hormone (GnRH) and GnRH receptor (GnRHR) in the hypothalamic-pituitary unit are reflected by luteinizing hormone (LH) secretion in sheep during anoestrous period and during luteal and follicular phases of the oestrous cycle. Using an enzyme-linked immunosorbent assay (ELISA), we analyzed the levels of GnRH and GnRHR in preoptic area (POA), anterior (AH) and ventromedial hypothalamus (VM), stalk-median eminence (SME), and GnRHR in the anterior pituitary gland (AP). Radioimmunoassay has also been used to define changes in plasma LH concentrations. The study provides evidence that the levels of GnRH in the whole hypothalamus of anoestrous ewes were lower than that in sheep during the follicular phase of the oestrous cycle (POA: p < 0.001, AH: p < 0.001, VM: p < 0.01, SME: p < 0.001) and not always than in luteal phase animals (POA: p < 0.05, SME: p < 0.05). It has also been demonstrated that the GnRHR amount in the hypothalamus-anterior pituitary unit, as well as LH level, in the blood in anoestrous ewes were significantly lower than those detected in animals of both cyclic groups. Our data suggest that decrease in LH secretion during the long photoperiod in sheep may be due to low translational activity of genes encoding both GnRH and GnRHR.


Asunto(s)
Anestro/metabolismo , Ciclo Estral/metabolismo , Hormona Liberadora de Gonadotropina/biosíntesis , Sistema Hipotálamo-Hipofisario/metabolismo , Receptores LHRH/biosíntesis , Anestro/sangre , Animales , Núcleo Hipotalámico Anterior/metabolismo , Ciclo Estral/sangre , Femenino , Hormona Luteinizante/sangre , Eminencia Media/metabolismo , Hipófisis/metabolismo , Área Preóptica/metabolismo , Ovinos , Núcleo Hipotalámico Ventromedial/metabolismo
7.
Bioorg Med Chem Lett ; 24(7): 1846-50, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24613701

RESUMEN

Two novel small molecule gonadotropin-releasing hormone (GnRH) receptor antagonists (12 and 13) of the furamide-class were synthesized and evaluated in vitro for their receptor binding affinities for the rat GnRH receptor. Radiolabeling with no carrier added fluorine-18 of the appropriate precursors was investigated in a one-step reaction. LogP (Octanol/PBS pH 7.4) and serum stability of the compounds were investigated. The antagonists showed low nM affinity for the rat GnRH receptor. (18)F-radiolabled compounds were obtained in high radiochemical purity (>95%) and specific activity (>75 GBq/µmol). These findings suggest this class of compounds holds promise as potential probes for PET targeting of GnRH-receptor expression.


Asunto(s)
Tomografía de Emisión de Positrones , Radiofármacos/farmacología , Receptores LHRH/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Relación Dosis-Respuesta a Droga , Radioisótopos de Flúor/química , Células HEK293 , Humanos , Estructura Molecular , Radiofármacos/síntesis química , Radiofármacos/química , Ratas , Receptores LHRH/biosíntesis , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad
8.
Int J Gynecol Cancer ; 24(2): 260-5, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24418927

RESUMEN

OBJECTIVE: Advanced or recurrent endometrial cancer (EC) no longer amenable to surgery or radiotherapy is a life-threatening disease with limited therapeutic options left. Eighty percent of ECs express receptors for luteinizing hormone-releasing hormone (LHRH), which can be targeted by AEZS-108 (zoptarelin doxorubicin acetate). This phase 2 trial was performed to assess the efficacy and safety of AEZS-108 in this group of patients. METHODS: Patients had FIGO (Fédération Internationale de Gynécologie et d'Obstétrique) III or IV or recurrent EC, LHRH receptor-positive tumor status, and at least had 1 measurable lesion (Response Evaluation Criteria in Solid Tumors). Prior anthracycline therapy was not allowed. Patients received AEZS-108 as a 2-hour infusion on day 1 of a 21-day cycle. The treatment was continued for a maximum of 6 to 8 cycles. The primary end point was the response rate determined by the Response Evaluation Criteria in Solid Tumors. RESULTS: From April 2008 to November 2009, 44 patients were included in the study at 8 centers in Germany (AGO) and 3 centers in Bulgaria. Forty-three of these patients were eligible. Two (5%) patients had a complete remission, and 8 (18%) achieved a partial remission. Stable disease for at least 6 weeks was observed in 44%. The median time to progression was 7 months, and the median overall survival was 15 months. The most frequently reported grade 3 or 4 adverse effects were neutropenia (12%) and leucopenia (9%). CONCLUSIONS: AEZS-108, an LHRH-agonist coupled to doxorubicin, has significant activity and low toxicity in women with advanced or recurrent LHRH receptor-positive EC, supporting the principle of receptor-mediated targeted chemotherapy.


Asunto(s)
Doxorrubicina/análogos & derivados , Neoplasias Endometriales/tratamiento farmacológico , Hormona Liberadora de Gonadotropina/análogos & derivados , Recurrencia Local de Neoplasia/tratamiento farmacológico , Receptores LHRH/antagonistas & inhibidores , Adulto , Anciano , Anciano de 80 o más Años , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Neoplasias Endometriales/metabolismo , Femenino , Hormona Liberadora de Gonadotropina/farmacología , Hormona Liberadora de Gonadotropina/uso terapéutico , Humanos , Persona de Mediana Edad , Recurrencia Local de Neoplasia/metabolismo , Receptores LHRH/biosíntesis
9.
Gen Comp Endocrinol ; 185: 57-66, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23395683

RESUMEN

To determine whether hormone-receptor signaling pathways in the thymus are altered by active immunization against gonadotrophin-releasing hormone I (GnRH), 3-week-old Sprague-Dawley male rats received GnRH-tandem-OVA peptides (200 µg/ml), and the effects were compared to a control group. Serum testosterone, LH and FSH concentrations were markedly reduced, with severe testicular atrophy, compared to controls, demonstrating effective blockade of the pituitary-gonadal axis. The reduction in LH and FSH concentrations in the thymus of immunized animals was lower than that observed in the serum, where a significant difference (P<0.001) in concentration was observed between both groups. Concentrations of GnRH were increased in the thymus of immunized rats. In thymic tissue, GnRHR, FSHR and LHR demonstrated stronger immunostaining, and AR weaker staining, in the immunized group compared to controls. Reproductive hormone receptor mRNA expression was consistent with protein variations in the immunized thymus. Compared to controls, GnRHR gene levels were significantly increased (P<0.05), however, AR mRNA expression were greatly decreased with immune week-age (P<0.05). Both FSHR and LHR mRNA expression levels were significantly higher in the treated group than in controls in the first three samples (P<0.05). When GnRHR was blocked by an antagonist in thymocytes, all reproductive hormone receptor gene expressions were significantly increased (P<0.001). In summary, these findings suggest that active immunization against GnRH can up-regulate GnRH receptor and gonadotropin receptor signaling, by stimulating thymic autocrine and paracrine function, whereas the androgen receptor is down-regulated due to a lack of testosterone secretion in the thymus.


Asunto(s)
Hormona Liberadora de Gonadotropina/inmunología , Receptores de HFE/biosíntesis , Receptores LHRH/biosíntesis , Receptores de HL/biosíntesis , Timo/metabolismo , Animales , Hormona Folículo Estimulante/sangre , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Hormona Luteinizante/sangre , Masculino , Ratas , Ratas Sprague-Dawley , Receptores de HFE/genética , Receptores de HL/genética , Receptores LHRH/genética , Testículo/inmunología , Testosterona/sangre , Timo/inmunología , Vacunación
10.
Int J Mol Sci ; 14(4): 7603-16, 2013 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-23567273

RESUMEN

Several evidences supported the existence of melatonin effects on reproductive system in fish. In order to investigate whether melatonin is involved in the modulation of GnRH systems in the European sea bass, we have injected melatonin (0.5 µg/g body mass) in male specimens. The brain mRNA transcript levels of the three GnRH forms and the five GnRH receptors present in this species were determined by real time quantitative PCR. Our findings revealed day-night variations in the brain expression of GnRH-1, GnRH-3 and several GnRH receptors (dlGnRHR-II-1c, -2a), which exhibited higher transcript levels at mid-light compared to mid-dark phase of the photocycle. Moreover, an inhibitory effect of melatonin on the nocturnal expression of GnRH-1, GnRH-3, and GnRH receptors subtypes 1c, 2a and 2b was also demonstrated. Interestingly, the inhibitory effect of melatonin affected the expression of hypophysiotrophic GnRH forms and GnRH receptors that exhibit day-night fluctuations, suggesting that exogenous melatonin reinforce physiological mechanisms already established. These interactions between melatoninergic and GnRH systems could be mediating photoperiod effects on reproductive and other rhythmic physiological events in the European sea bass.


Asunto(s)
Lubina/metabolismo , Encéfalo/metabolismo , Proteínas de Peces/biosíntesis , Regulación de la Expresión Génica/fisiología , Melatonina/metabolismo , Receptores LHRH/biosíntesis , Animales , Femenino , Masculino , Fotoperiodo , Reproducción/fisiología
11.
Am J Physiol Endocrinol Metab ; 303(4): E475-87, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22669247

RESUMEN

Gonadotropin-releasing hormone (either GnRH1 or GnRH2) exerts a local activity in vertebrate testis, including human testis. Relationships between endocannabinoid (eCB) and GnRH systems in gonads have never been elucidated in any species so far. To reveal a cross-talk between eCBs and GnRH at testicular level, we characterized the expression of GnRH (GnRH1 and GnRH2) as well as GnRH receptor (GnRH-R1, -R2, and -R3) mRNA in the testis of the anuran amphibian Rana esculenta during the annual sexual cycle; furthermore, the corresponding transcripts were localized inside the testis by in situ hybridization. The possible endogenous production of the eCB, anandamide (AEA), was investigated in testis by analyzing the expression of its biosynthetic enzyme, Nape-pld. Incubations of testis pieces with AEA were carried out in the postreproductive period (June) and in February, when a new spermatogenetic wave takes place. In June, AEA treatment significantly decreased GnRH1 and GnRH-R2 mRNA, stimulated the transcription of GnRH2 and GnRH-R1, and did not affect GnRH-R3 expression. In February, AEA treatment upregulated GnRH2 and GnRH-R3 mRNA, downregulated GnRH-R2, and did not affect GnRH1 and GnRH-R1 expression. These effects were mediated by type 1 cannabinoid receptor (CB1) since they were fully counteracted by SR141716A (Rimonabant), a selective CB1 antagonist. In conclusion, eCB system modulates GnRH activity in frog testis during the annual sexual cycle in a stage-dependent fashion.


Asunto(s)
Ácidos Araquidónicos/farmacología , Endocannabinoides/farmacología , Hormona Liberadora de Gonadotropina/metabolismo , Alcamidas Poliinsaturadas/farmacología , Receptores LHRH/biosíntesis , Testículo/efectos de los fármacos , Testículo/metabolismo , Animales , Ácidos Araquidónicos/biosíntesis , Endocannabinoides/biosíntesis , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Masculino , Fosfolipasa D/metabolismo , Piperidinas/farmacología , Pirazoles/farmacología , Rana esculenta , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB1/metabolismo , Receptores LHRH/antagonistas & inhibidores , Rimonabant , Estaciones del Año , Espermatogénesis/efectos de los fármacos , Espermatogénesis/fisiología
12.
Horm Behav ; 62(1): 18-26, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22561338

RESUMEN

Feeding behavior and reproduction are coordinately regulated by the brain via neurotransmitters, circulating hormones, and neuropeptides. Reduced feeding allows animals to engage in other behaviors important for fitness, including mating and parental care. Some fishes cease feeding for weeks at a time in order to provide care to their young by brooding them inside the male or female parent's mouth. Maternal mouthbrooding is known to impact circulating hormones and subsequent reproductive cycles, but neither the full effects of food deprivation nor the neural mechanisms are known. Here we ask what effects mouthbrooding has on several physiological processes including gonad and body mass, brain neuropeptide and receptor gene expression, and circulating steroid hormones in a mouthbrooding cichlid species, Astatotilapia burtoni. We ask whether any observed changes can be explained by food deprivation, and show that during mouthbrooding, ovary size and circulating levels of androgens and estrogens match those seen during food deprivation. Levels of gonadotropin-releasing hormone 1 (GnRH1) mRNA in the brain were low in food-deprived females compared to controls and in mouthbrooding females compared to gravid females. Levels of mRNA encoding two peptides involved in regulating feeding, hypocretin and cholecystokinin, were increased in the brains of food-deprived females. Brain mRNA levels of two receptors, GnRH receptor 2 and NPY receptor Y8c, were elevated in mouthbrooding females compared to the fed condition, but NPY receptor Y8b mRNA was differently regulated by mouthbrooding. These results suggest that many, but not all, of the characteristic physiological changes that occur during mouthbrooding are consequences of food deprivation.


Asunto(s)
Andrógenos/sangre , Cíclidos/metabolismo , Estrógenos/sangre , Privación de Alimentos/fisiología , Neuropéptidos/metabolismo , Ovario/metabolismo , Animales , Peso Corporal , Encéfalo/metabolismo , Colecistoquinina/biosíntesis , Femenino , Hormona Liberadora de Gonadotropina/biosíntesis , Hormona Liberadora de Gonadotropina/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Neuropéptidos/biosíntesis , Orexinas , Tamaño de los Órganos , Ovario/anatomía & histología , Precursores de Proteínas/biosíntesis , ARN Mensajero/biosíntesis , Receptores LHRH/biosíntesis , Receptores de Neuropéptido Y/biosíntesis , Reproducción
13.
Anticancer Drugs ; 23(9): 959-69, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22713594

RESUMEN

Gonadotropin-releasing hormone (GnRH) agonists are widely used for the treatment of advanced prostate cancer (PCa). Agonists activate the GnRH receptor (GnRH-R), triggering apoptosis in PCa cells. In gonadotropes, the amount of GnRH-R in the plasma membrane is regulated by protein folding and endoplasmic reticulum retention, mechanisms that can be overcome by the pharmacoperone IN3. Our aim was to describe the intracellular distribution of GnRH-R in PCa cells and its relation to response to GnRH analog treatments. The expressions of GnRH-R in PCa biopsies were evaluated by immunohistochemistry and the intracellular distribution was determined by immunofluorescence in primary cell cultures from human PCa samples. Cultured cells were pretreated with IN3 and then with leuprolide. Cell survival was evaluated by 1-(4,5-dimethylthiazol-2-yl)-3,5-diphenylformazan (MTT) thiazolyl blue formazan and cell cycle and apoptosis by flow cytometry. We observed that the expression of GnRH-R decreased according to malignant progression. Most GnRH-R are located inside the cell, colocalizing with endoplasmic reticulum markers. The treatment with IN3 decreased cellular GnRH-R retention, increasing plasma membrane expression in approximately 60%. Pretreatment with IN3 decreased PCa cell survival compared with leuprolide-alone treatment, primarily because of an increase in apoptosis. We conclude that the response of PCa cells to leuprolide is related to the amount of GnRH-R in the plasma membrane. Therefore, pretreatment evaluation of the amount of these receptors may be a predictor of the outcome of leuprolide treatment in PCa patients. Assessment of systemic IN3 effect would be necessary to determine its utility as an adjuvant treatment in hormone-resistant tumors.


Asunto(s)
Apoptosis/efectos de los fármacos , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Membrana Celular/efectos de los fármacos , Indoles/farmacología , Leuprolida/farmacología , Neoplasias de la Próstata/patología , Piridinas/farmacología , Receptores LHRH/agonistas , Western Blotting , Técnicas de Cultivo de Célula , Ciclo Celular/efectos de los fármacos , Membrana Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Citometría de Flujo , Humanos , Inmunohistoquímica , Masculino , Neoplasias de la Próstata/metabolismo , Pliegue de Proteína , Receptores LHRH/biosíntesis , Células Tumorales Cultivadas
14.
Yao Xue Xue Bao ; 47(4): 452-8, 2012 Apr.
Artículo en Zh | MEDLINE | ID: mdl-22799026

RESUMEN

This study is to investigate the anti-tumor effect in vitro of methotrexate modified by LH-RH peptide (LH-RH-MTX). LH-RH receptors highly expressing MCF-7 human breast carcinoma cell line and lowly expressing K562 human erythroleukemia cell line were served as the tested cells. The cell proliferation inhibition rates of LH-RH-MTX were detected by MTT colorimetric assay. The effects of LH-RH-MTX on the cell cycle and apoptosis rates were detected by flow cytometry. The inhibition rate of LH-RH-MTX on MCF-7 cells was much higher than that on K562 cells, and the inhibition rate of LH-RH-MTX on MCF-7 cells was much higher than that of free MTX at the same concentration. The inhibition rate of LH-RH-MTX on rat bone marrow mononuclear cells was less than that of free MTX. The number of MCF-7 cells in S phase increased after administration of LH-RH-MTX. The apoptosis rate of LH-RH-MTX group significantly increased compared with that of the control group and MTX group. The relative expression of LHRHR mRNA of LH-RH-MTX group markedly decreased compared with that of the control group and MTX group. LH-RH-MTX is realizable to reduce drug side effects, increase the therapeutic index and achieve tumor-targeted therapy.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Metotrexato/farmacología , Receptores LHRH/biosíntesis , Animales , Antimetabolitos Antineoplásicos/síntesis química , Apoptosis/efectos de los fármacos , Células de la Médula Ósea/citología , Ciclo Celular/efectos de los fármacos , Células Cultivadas , Hormona Liberadora de Gonadotropina/química , Hormona Liberadora de Gonadotropina/farmacología , Humanos , Células K562 , Leucocitos Mononucleares , Células MCF-7 , Metotrexato/síntesis química , ARN Mensajero/metabolismo , Ratas , Receptores LHRH/genética
15.
Prostate ; 71(5): 445-52, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20859992

RESUMEN

BACKGROUND: The majority of men will develop symptoms of benign prostatic hyperplasia (BPH) after 70 years of age. Various studies indicate that antagonists of LHRH, such as cetrorelix, exert direct inhibitory effects on BPH mediated by specific LHRH receptors. Our aim was to investigate the mRNA for LHRH and LHRH receptors and the expression of LHRH receptors in specimens of human BPH. METHODS: The expression of mRNA for LHRH (n=35) and LHRH receptors (n=55) was investigated by RT-PCR in surgical specimens of BPH, using specific primers. The characteristics of binding sites for LHRH on 20 samples were determined by ligand competition assays. The LHRH receptor expression was also examined in 64 BPH specimens by immunohistochemistry. RESULTS: PCR products for LHRH were found in 18 of 35 (51%) BPH tissues and mRNA for LHRH receptors was detected in 39 of 55 (71%) BPH specimens. Eighteen of 20 (90%) samples showed a single class of high affinity binding sites for [D-Trp(6) ]LHRH with a mean K(d) of 4.04 nM and a mean B(max) of 527.6 fmol/mg membrane protein. LHRH antagonist cetrorelix showed high affinity binding to LHRH receptors in BPH. Positive immunohistochemical reaction for LHRH receptors was present in 42 of 64 (67%) BPH specimens. CONCLUSION: A high incidence of LHRH receptors in BPH supports the use of LHRH antagonists such as cetrorelix, for treatment of patients with lower urinary tract symptoms from BPH.


Asunto(s)
Hormona Liberadora de Gonadotropina/análogos & derivados , Antagonistas de Hormonas/farmacología , Hiperplasia Prostática/metabolismo , Receptores LHRH/biosíntesis , Anciano , Anciano de 80 o más Años , Unión Competitiva , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Liberadora de Gonadotropina/farmacología , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Terapia Molecular Dirigida/métodos , Hiperplasia Prostática/tratamiento farmacológico , Hiperplasia Prostática/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ensayo de Unión Radioligante , Receptores LHRH/antagonistas & inhibidores , Receptores LHRH/genética , Receptores LHRH/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Prostate ; 71(9): 915-28, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21541969

RESUMEN

BACKGROUND AND AIMS: Human metastatic prostate cancer cell growth can be inhibited by GnRH analogs but effects on virus-immortalized prostate cells have not been investigated. METHODS: Virus-immortalized prostate cells were stably transfected with rat GnRH receptor cDNA and levels of GnRH binding were correlated with GnRH effects on signaling, cell cycle, growth, exosome production, and apoptosis. RESULTS: High levels of cell surface GnRH receptor occurred in transfected papillomavirus-immortalized WPE-1-NB26 epithelial cells but not in non-tumourigenic RWPE-1, myoepithelial WPMY-1 cells, or SV40-immortalized PNT1A. Endogenous cell surface GnRH receptor was undetectable in non-transfected cells or cancer cell lines LNCaP, PC3, and DU145. GnRH receptor levels correlated with induction of inositol phosphates, elevation of intracellular Ca(2+) , cytoskeletal actin reorganization, modulation of ERK activation and cell growth-inhibition with GnRH agonists. Hoechst 33342 DNA staining-cell sorting indicated accumulation of cells in G2 following agonist treatment. Release of exosomes from transfected WPE-1-NB26 was unaffected by agonists, unlike induction observed in HEK293([SCL60]) cells. Increased PARP cleavage and apoptotic body production were undetectable during growth-inhibition in WPE-1-NB26 cells, contrasting with HEK293([SCL60]) . EGF receptor activation inhibited GnRH-induced ERK activation in WPE-1-NB26 but growth-inhibition was not rescued by EGF or PKC inhibitor Ro320432. Growth of cells expressing low levels of GnRH receptor was not affected by agonists. CONCLUSIONS: Engineered high-level GnRH receptor activation inhibits growth of a subset of papillomavirus-immortalized prostate cells. Elucidating mechanisms leading to clone-specific differences in cell surface GnRH receptor levels is a valuable next step in developing strategies to exploit prostate cell anti-proliferation using GnRH agonists.


Asunto(s)
Hormona Liberadora de Gonadotropina/agonistas , Neoplasias de la Próstata/metabolismo , Receptores LHRH/biosíntesis , Pamoato de Triptorelina/farmacología , Alphapapillomavirus/fisiología , Apoptosis/fisiología , Ciclo Celular/fisiología , Línea Celular Tumoral , Transformación Celular Viral , Exosomas/fisiología , Citometría de Flujo , Hormona Liberadora de Gonadotropina/metabolismo , Humanos , Fosfatos de Inositol/metabolismo , Masculino , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/virología , Receptores LHRH/genética , Receptores LHRH/metabolismo , Transducción de Señal/fisiología , Transfección
17.
Hum Reprod ; 25(11): 2878-90, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20829343

RESUMEN

BACKGROUND: We recently demonstrated the effect of gonadotrophin-releasing hormone agonist (GnRHa) on tissue inflammation, angiogenesis and apoptosis in endometriosis, adenomyosis and uterine myoma. Here, we investigated expression of GnRH receptors (GnRHRs) and effect of GnRHa on the proliferation of cells derived from endometria and pathological lesions of women with these reproductive diseases. METHODS: Biopsy specimens were collected from lesions and corresponding endometria of 35 women with pelvic endometriosis, 45 women with ovarian endometrioma, 35 women with adenomyosis and 56 women with uterine myoma during laparoscopy or laparotomy. The gene and protein expressions of GnRHR in eutopic/ectopic cells and tissues were examined by reverse transcriptase-polymerase chain reaction (RT-PCR) and immunohistochemistry. The immunoreactivity of GnRHR in tissue was analysed by quantitative-histogram (Q-H) scores. The exogenous effect of GnRHa on cell proliferation was examined by 5-bromo-2-deoxyuridine incorporation assay. The Ki-67-immunoreactive cell proliferation index was analysed in biopsy specimens derived from GnRHa-treated and -non-treated women. RESULTS: Types I and II GnRHRs mRNA and proteins were expressed in eutopic endometria and pathological lesions derived from women with endometriosis, adenomyosis and uterine myoma. GnRHR expression was the highest in the menstrual phase when compared with other phases of the menstrual cycle. Higher Q-H scores of GnRHR immunoreaction were found in blood-filled opaque red lesions than in other peritoneal lesions. Exogenous treatment with GnRHa significantly suppressed the proliferation of cells derived from respective endometria and pathological lesions when compared with GnRHa-non-treated cells. CONCLUSIONS: Local tissue expression of GnRHR was detected in endometriosis, adenomyosis and uterine myoma. In addition to a hypo-estrogenic effect, a direct anti-proliferative effect of GnRHa may be involved in the regression of these reproductive diseases with consequent remission of clinical symptoms.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Endometriosis/patología , Hormona Liberadora de Gonadotropina/agonistas , Leiomioma/patología , Leuprolida/farmacología , Neoplasias Ováricas/patología , Receptores LHRH/biosíntesis , Neoplasias Uterinas/patología , Adulto , Endometriosis/metabolismo , Femenino , Humanos , Antígeno Ki-67/biosíntesis , Leiomioma/metabolismo , Neoplasias Ováricas/metabolismo , Neoplasias Uterinas/metabolismo
18.
Gynecol Oncol ; 119(3): 457-61, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20828803

RESUMEN

OBJECTIVES: Receptors for luteinizing hormone-releasing hormone (LHRH) can be utilized for targeted chemotherapy of cytotoxic LHRH analogs. The compound AEZS-108 (previously AN-152) consists of [D-Lys6]LHRH linked to doxorubicin. The objectives of this first study in humans with AESZ-108 were to determine the maximum tolerated dose and to characterize the dose-limiting toxicity, pharmacokinetics, preliminary efficacy, and hormonal effects. METHODS: The study included 17 women with histologically confirmed epithelial cancer of the ovary, endometrium, or breast that was metastatic or unresectable and for which standard curative or palliative measures could not be used or were no longer effective or tolerated. In each patient, immunohistochemistry of primary tumor or metastatic lesion confirmed that the tumors expressed LHRH receptors. RESULTS: One patient each received intravenous doses of 10, 20, 40, or 80 mg/m² of AEZS-108, six received 160 mg/m² and seven 267 mg/m² at 3 week intervals. Dose-limiting leukopenia and neutropenia were observed at the highest dose. A total of 6 patients, 3 patients each in both upper dose groups, showed responses to AEZS-108. The half-life of AESZ-108 was estimated to be about 2h. CONCLUSIONS: The maximum tolerated dose of AESZ-108 in the absence of supportive medication is 267 mg/m² and this dose is recommended as starting dose for therapeutic Phase II studies.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Doxorrubicina/análogos & derivados , Neoplasias Endometriales/tratamiento farmacológico , Hormona Liberadora de Gonadotropina/análogos & derivados , Hormona Liberadora de Gonadotropina/agonistas , Neoplasias Ováricas/tratamiento farmacológico , Receptores LHRH/biosíntesis , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/efectos adversos , Antibióticos Antineoplásicos/sangre , Antibióticos Antineoplásicos/farmacocinética , Neoplasias de la Mama/sangre , Neoplasias de la Mama/metabolismo , Relación Dosis-Respuesta a Droga , Doxorrubicina/administración & dosificación , Doxorrubicina/efectos adversos , Doxorrubicina/sangre , Doxorrubicina/farmacocinética , Neoplasias Endometriales/sangre , Neoplasias Endometriales/metabolismo , Femenino , Hormona Liberadora de Gonadotropina/administración & dosificación , Hormona Liberadora de Gonadotropina/efectos adversos , Hormona Liberadora de Gonadotropina/sangre , Hormona Liberadora de Gonadotropina/farmacocinética , Humanos , Hidrocortisona/metabolismo , Dosis Máxima Tolerada , Persona de Mediana Edad , Neoplasias Ováricas/sangre , Neoplasias Ováricas/metabolismo , Hipófisis/efectos de los fármacos
19.
J Neurochem ; 110(3): 1014-27, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19493163

RESUMEN

Brain sex steroids are derived from both peripheral (primarily gonadal) and local (neurosteroids) sources and are crucial for neurogenesis, neural differentiation and neural function. The mechanism(s) regulating the production of neurosteroids is not understood. To determine whether hypothalamic-pituitary-gonadal axis components previously detected in the extra-hypothalamic brain comprise a feedback loop to regulate neuro-sex steroid (NSS) production, we assessed dynamic changes in expression patterns of steroidogenic acute regulatory (StAR) protein, a key regulator of steroidogenesis, and key hypothalamic-pituitary-gonadal endocrine receptors, by modulating peripheral sex hormone levels in female mice. Ovariectomy (OVX; high serum gonadotropins, low serum sex steroids) had a differential effect on StAR protein levels in the extrahypothalamic brain; increasing the 30- and 32-kDa variants but decreasing the 37-kDa variant and is indicative of cholesterol transport into mitochondria for steroidogenesis. Treatment of OVX animals with E(2), P(4), or E(2) + P(4) for 3 days, which decreases OVX-induced increases in GnRH/gonadotropin production, reversed this pattern. Suppression of gonadotropin levels in OVX mice using the GnRH agonist leuprolide acetate inhibited the processing of the 37-kDa StAR protein into the 30-kDa StAR protein, confirming that the differential processing of brain StAR protein is regulated by gonadotropins. OVX dramatically suppressed extra-hypothalamic brain gonadotropin-releasing hormone 1 receptor expression, and was further suppressed in E(2)- or P(4)-treated OVX mice. Together, these data indicate the existence of endocrine and autocrine/paracrine feedback loops that regulate NSS synthesis. Further delineation of these feedback loops that regulate NSS production will aid in developing therapies to maintain brain sex steroid levels and cognition.


Asunto(s)
Hormonas Esteroides Gonadales/biosíntesis , Sistema Hipotálamo-Hipofisario/metabolismo , Ovario/metabolismo , Fosfoproteínas/biosíntesis , Receptores LHRH/biosíntesis , Animales , Retroalimentación Fisiológica/fisiología , Femenino , Humanos , Hipotálamo/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Neurotransmisores/biosíntesis , Hipófisis/metabolismo
20.
Oncol Rep ; 21(5): 1277-82, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19360304

RESUMEN

Malignant glioblastoma is one of the highest proliferative and invasive tumors within the central nervous system (CNS); the therapeutical options for this disease are still very poor. Receptors for gonadotropin-releasing hormone (GnRH) have been reported to be present in glioblastoma tissues. This study aimed to determine the role of these receptors in the control of glioma growth. In two human glioblastoma cell lines, U87MG and U373, we demonstrated the expression of GnRH receptors, both at mRNA and protein levels. We also found that GnRH receptor is expressed in glioblastoma tissues from tumor patients as shown by Western blotting. In U87MG and U373 cell lines, we found the expression of mRNA for GnRH, indicating the presence of an autocrine GnRH-based system in these cell lines. Treatment of the two cell lines with a GnRH agonist resulted in a significant decrease of cell proliferation. Moreover, the GnRH agonist significantly counteracted the forskolin-induced increase of intracellular cAMP levels in these cells. These findings suggest that the GnRH receptor might represent a molecular target for an endocrine therapeutical approach against gliomas.


Asunto(s)
Glioblastoma/metabolismo , Hormona Liberadora de Gonadotropina/biosíntesis , Receptores LHRH/biosíntesis , Antineoplásicos Hormonales/farmacología , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , AMP Cíclico/metabolismo , Glioblastoma/genética , Glioblastoma/patología , Hormona Liberadora de Gonadotropina/agonistas , Hormona Liberadora de Gonadotropina/genética , Goserelina/farmacología , Humanos , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores LHRH/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA