Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
FASEB J ; 35(4): e21464, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33724574

RESUMEN

Chemical castration in prostate cancer can be achieved with gonadotropin-releasing hormone (GnRH) agonists or antagonists. Their effects differ by the initial flare of gonadotropin and testosterone secretion with agonists and the immediate pituitary-testicular suppression by antagonists. While both suppress luteinizing hormone (LH) and follicle-stimulating hormone (FSH) initially, a rebound in FSH levels occurs during agonist treatment. This rebound is potentially harmful, taken the expression of FSH receptors (R) in prostate cancer tissue. We herein assessed the role of FSH in promoting the growth of androgen-independent (PC-3, DU145) and androgen-dependent (VCaP) human prostate cancer cell line xenografts in nude mice. Gonadotropins were suppressed with the GnRH antagonist degarelix, and effects of add-back human recombinant FSH were assessed on tumor growth. All tumors expressed GnRHR and FSHR, and degarelix treatment suppressed their growth. FSH supplementation reversed the degarelix-evoked suppression of PC-3 tumors, both in preventive (degarelix and FSH treatment started upon cell inoculation) and therapeutic (treatments initiated 3 weeks after cell inoculation) setting. A less marked, though significant FSH effect occurred in DU145, but not in VCaP xenografts. FSHR expression in the xenografts supports direct FSH stimulation of tumor growth. Testosterone supplementation, to maintain the VCaP xenografts, apparently masked the FSH effect on their growth. Treatment with the LH analogue hCG did not affect PC-3 tumor growth despite their expression of luteinizing hormone/choriongonadotropin receptor. In conclusion, FSH, but not LH, may directly stimulate the growth of androgen-independent prostate cancer, suggesting that persistent FSH suppression upon GnRH antagonist treatment offers a therapeutic advantage over agonist.


Asunto(s)
Hormona Folículo Estimulante/farmacología , Xenoinjertos/efectos de los fármacos , Hormona Luteinizante/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Andrógenos/farmacología , Animales , Línea Celular , Hormona Folículo Estimulante/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Hormona Liberadora de Gonadotropina/farmacología , Humanos , Masculino , Ratones Desnudos , Neoplasias de la Próstata/metabolismo , Receptores de HFE , Testículo/metabolismo , Testosterona/farmacología
3.
J Clin Endocrinol Metab ; 103(9): 3420-3429, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29931354

RESUMEN

Context: Self-limited delayed puberty (DP) segregates in an autosomal-dominant pattern, but the genetic basis is largely unknown. Although DP is sometimes seen in relatives of patients with hypogonadotropic hypogonadism (HH), mutations in genes known to cause HH that segregate with the trait of familial self-limited DP have not yet been identified. Objective: To assess the contribution of mutations in genes known to cause HH to the phenotype of self-limited DP. Design, Patients, and Setting: We performed whole-exome sequencing in 67 probands and 93 relatives from a large cohort of familial self-limited DP, validated the pathogenicity of the identified gene variant in vitro, and examined the tissue expression and functional requirement of the mouse homolog in vivo. Results: A potentially pathogenic gene variant segregating with DP was identified in 1 of 28 known HH genes examined. This pathogenic variant occurred in HS6ST1 in one pedigree and segregated with the trait in the six affected members with heterozygous transmission (P = 3.01 × 10-5). Biochemical analysis showed that this mutation reduced sulfotransferase activity in vitro. Hs6st1 mRNA was expressed in peripubertal wild-type mouse hypothalamus. GnRH neuron counts were similar in Hs6st1+/- and Hs6st1+/+ mice, but vaginal opening was delayed in Hs6st1+/- mice despite normal postnatal growth. Conclusions: We have linked a deleterious mutation in HS6ST1 to familial self-limited DP and show that heterozygous Hs6st1 loss causes DP in mice. In this study, the observed overlap in potentially pathogenic mutations contributing to the phenotypes of self-limited DP and HH was limited to this one gene.


Asunto(s)
Hipogonadismo/genética , Pubertad Tardía/genética , Sulfotransferasas/deficiencia , Animales , Estudios de Cohortes , Femenino , Finlandia , Hormona Liberadora de Gonadotropina/genética , Heterocigoto , Humanos , Hipotálamo/metabolismo , Masculino , Ratones , Mutación , Linaje , Fenotipo , Sulfotransferasas/genética , Secuenciación del Exoma
4.
J Clin Invest ; 128(5): 1787-1792, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29584617

RESUMEN

Spermatogenesis is regulated by the 2 pituitary gonadotropins, luteinizing hormone (LH) and follicle-stimulating hormone (FSH). This process is considered impossible without the absolute requirement of LH-stimulated testicular testosterone (T) production. The role of FSH remains unclear because men and mice with inactivating FSH receptor (FSHR) mutations are fertile. We revisited the role of FSH in spermatogenesis using transgenic mice expressing a constitutively strongly active FSHR mutant in a LH receptor-null (LHR-null) background. The mutant FSHR reversed the azoospermia and partially restored fertility of Lhr-/- mice. The finding was initially ascribed to the residual Leydig cell T production. However, when T action was completely blocked with the potent antiandrogen flutamide, spermatogenesis persisted. Hence, completely T-independent spermatogenesis is possible through strong FSHR activation, and the dogma of T being a sine qua non for spermatogenesis may need modification. The mechanism for the finding appeared to be that FSHR activation maintained the expression of Sertoli cell genes considered androgen dependent. The translational message of our findings is the possibility of developing a new strategy of high-dose FSH treatment for spermatogenic failure. Our findings also provide an explanation of molecular pathogenesis for Pasqualini syndrome (fertile eunuchs; LH/T deficiency with persistent spermatogenesis) and explain how the hormonal regulation of spermatogenesis has shifted from FSH to T dominance during evolution.


Asunto(s)
Hormona Folículo Estimulante/metabolismo , Células Intersticiales del Testículo/metabolismo , Receptores de HFE/metabolismo , Células de Sertoli/metabolismo , Espermatogénesis , Andrógenos/genética , Andrógenos/metabolismo , Animales , Hormona Folículo Estimulante/genética , Masculino , Ratones , Ratones Transgénicos , Receptores de HFE/genética , Receptores de HL/metabolismo , Testosterona/genética , Testosterona/metabolismo
5.
J Clin Endocrinol Metab ; 103(2): 649-659, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29161441

RESUMEN

Context: Self-limited delayed puberty (DP) is often associated with a delay in physical maturation, but although highly heritable the causal genetic factors remain elusive. Genome-wide association studies of the timing of puberty have identified multiple loci for age at menarche in females and voice break in males, particularly in pathways controlling energy balance. Objective/Main Outcome Measures: We sought to assess the contribution of rare variants in such genes to the phenotype of familial DP. Design/Patients: We performed whole-exome sequencing in 67 pedigrees (125 individuals with DP and 35 unaffected controls) from our unique cohort of familial self-limited DP. Using a whole-exome sequencing filtering pipeline one candidate gene [fat mass and obesity-associated gene (FTO)] was identified. In silico, in vitro, and mouse model studies were performed to investigate the pathogenicity of FTO variants and timing of puberty in FTO+/- mice. Results: We identified potentially pathogenic, rare variants in genes in linkage disequilibrium with genome-wide association studies of age at menarche loci in 283 genes. Of these, five genes were implicated in the control of body mass. After filtering for segregation with trait, one candidate, FTO, was retained. Two FTO variants, found in 14 affected individuals from three families, were also associated with leanness in these patients with DP. One variant (p.Leu44Val) demonstrated altered demethylation activity of the mutant protein in vitro. Fto+/- mice displayed a significantly delayed timing of pubertal onset (P < 0.05). Conclusions: Mutations in genes implicated in body mass and timing of puberty in the general population may contribute to the pathogenesis of self-limited DP.


Asunto(s)
Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/genética , Peso Corporal/genética , Polimorfismo de Nucleótido Simple , Pubertad Tardía/genética , Pubertad/genética , Adolescente , Animales , Índice de Masa Corporal , Estudios de Casos y Controles , Niño , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Ratones , Ratones Noqueados , Linaje , Fenotipo , Factores de Tiempo
6.
Stem Cell Res ; 25: 233-244, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29172153

RESUMEN

The class Ia anti-arrhythmic drug ajmaline is used clinically to unmask latent type I ECG in Brugada syndrome (BrS) patients, although its mode of action is poorly characterised. Our aims were to identify ajmaline's mode of action in human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CMs), and establish a simple BrS hiPSC platform to test whether differences in ajmaline response could be determined between BrS patients and controls. Control hiPSCs were differentiated into spontaneously contracting cardiac clusters. It was found using multi electrode array (MEA) that ajmaline treatment significantly lengthened cluster activation-recovery interval. Patch clamping of single CMs isolated from clusters revealed that ajmaline can block both INa and IKr. Following generation of hiPSC lines from BrS patients (absent of pathogenic SCN5A sodium channel mutations), analysis of hiPSC-CMs from patients and controls revealed that differentiation and action potential parameters were similar. Comparison of cardiac clusters by MEA showed that ajmaline lengthened activation-recovery interval consistently across all lines. We conclude that ajmaline can block both depolarisation and repolarisation of hiPSC-CMs at the cellular level, but that a more refined integrated tissue model may be necessary to elicit differences in its effect between BrS patients and controls.


Asunto(s)
Ajmalina/administración & dosificación , Antiarrítmicos/administración & dosificación , Síndrome de Brugada/tratamiento farmacológico , Corazón/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Células Madre Pluripotentes/efectos de los fármacos , Adulto , Síndrome de Brugada/genética , Síndrome de Brugada/metabolismo , Síndrome de Brugada/fisiopatología , Diferenciación Celular/efectos de los fármacos , Corazón/fisiopatología , Humanos , Masculino , Persona de Mediana Edad , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Técnicas de Placa-Clamp , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo
7.
Mol Cell Endocrinol ; 447: 12-22, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28232089

RESUMEN

GnRH neurons are fundamental for reproduction in all vertebrates, integrating all reproductive inputs. The inaccessibility of human GnRH-neurons has been a major impediment to studying the central control of reproduction and its disorders. Here, we report the efficient generation of kisspeptin responsive GnRH-secreting neurons by directed differentiation of human Embryonic Stem Cells and induced-Pluripotent Stem Cells derived from a Kallman Syndrome patient and a healthy family member. The protocol involves the generation of intermediate Neural Progenitor Cells (NPCs) through long-term Bone morphogenetic protein 4 inhibition, followed by terminal specification of these NPCs in media containing Fibroblast Growth Factor 8 and a NOTCH inhibitor. The resulting GnRH-expressing and -secreting neurons display a neuroendocrine gene expression pattern and present spontaneous calcium transients that can be stimulated by kisspeptin. These in vitro generated GnRH expressing cells provide a new resource for studying the molecular mechanisms underlying the development and function of GnRH neurons.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Neuronas/metabolismo , Células Madre Pluripotentes/metabolismo , Calcio/metabolismo , Diferenciación Celular , Línea Celular , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Células Madre Embrionarias Humanas/metabolismo , Humanos , Células-Madre Neurales/metabolismo
8.
Cell Rep ; 16(4): 1026-1038, 2016 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-27396331

RESUMEN

A surface marker that distinctly identifies cardiac progenitors (CPs) is essential for the robust isolation of these cells, circumventing the necessity of genetic modification. Here, we demonstrate that a Glycosylphosphatidylinositol-anchor containing neurotrophic factor receptor, Glial cell line-derived neurotrophic factor receptor alpha 2 (Gfra2), specifically marks CPs. GFRA2 expression facilitates the isolation of CPs by fluorescence activated cell sorting from differentiating mouse and human pluripotent stem cells. Gfra2 mutants reveal an important role for GFRA2 in cardiomyocyte differentiation and development both in vitro and in vivo. Mechanistically, the cardiac GFRA2 signaling pathway is distinct from the canonical pathway dependent on the RET tyrosine kinase and its established ligands. Collectively, our findings establish a platform for investigating the biology of CPs as a foundation for future development of CP transplantation for treating heart failure.


Asunto(s)
Diferenciación Celular/fisiología , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas Proto-Oncogénicas c-ret/metabolismo , Transducción de Señal/fisiología , Animales , Células Cultivadas , Glicosilfosfatidilinositoles/metabolismo , Humanos , Ligandos , Ratones , Organogénesis/fisiología , Células Madre Pluripotentes/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo
9.
Cancer Res ; 74(19): 5469-79, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25145671

RESUMEN

RIP140 is a transcriptional coregulator involved in energy homeostasis, ovulation, and mammary gland development. Although conclusive evidence is lacking, reports have implicated a role for RIP140 in breast cancer. Here, we explored the mechanistic role of RIP140 in breast cancer and its involvement in estrogen receptor α (ERα) transcriptional regulation of gene expression. Using ChIP-seq analysis, we demonstrate that RIP140 shares more than 80% of its binding sites with ERα, colocalizing with its interaction partners FOXA1, GATA3, p300, CBP, and p160 family members at H3K4me1-demarcated enhancer regions. RIP140 is required for ERα-complex formation, ERα-mediated gene expression, and ERα-dependent breast cancer cell proliferation. Genes affected following RIP140 silencing could be used to stratify tamoxifen-treated breast cancer cohorts, based on clinical outcome. Importantly, this gene signature was only effective in endocrine-treated conditions. Cumulatively, our data suggest that RIP140 plays an important role in ERα-mediated transcriptional regulation in breast cancer and response to tamoxifen treatment.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Receptor alfa de Estrógeno/fisiología , Proteínas Nucleares/fisiología , Transcripción Genética/fisiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Línea Celular Tumoral , Cromatina/metabolismo , Humanos , Células MCF-7 , Proteínas Nucleares/metabolismo , Proteína de Interacción con Receptores Nucleares 1 , Unión Proteica
10.
Development ; 140(5): 1079-89, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23404106

RESUMEN

Nuclear receptor interacting protein (Nrip1), also known as RIP140, is a co-regulator for nuclear receptors that plays an essential role in ovulation by regulating the expression of the epidermal growth factor-like family of growth factors. Although several studies indicate a role for RIP140 in breast cancer, its role in the development of the mammary gland is unclear. By using RIP140-null and RIP140 transgenic mice, we demonstrate that RIP140 is an essential factor for normal mammary gland development and that it functions by mediating oestrogen signalling. RIP140-null mice exhibit minimal ductal elongation with no side-branching, whereas RIP140-overexpressing mice show increased cell proliferation and ductal branching with age. Tissue recombination experiments demonstrate that RIP140 expression is required in both the mammary epithelial and stromal compartments for ductal elongation during puberty and that loss of RIP140 leads to a catastrophic loss of the mammary epithelium, whereas RIP140 overexpression augments the mammary basal cell population and shifts the progenitor/differentiated cell balance within the luminal cell compartment towards the progenitors. For the first time, we present a genome-wide global view of oestrogen receptor-α (ERα) binding events in the developing mammary gland, which unravels 881 ERα binding sites. Unbiased evaluation of several ERα binding sites for RIP140 co-occupancy reveals selectivity and demonstrates that RIP140 acts as a co-regulator with ERα to regulate directly the expression of amphiregulin (Areg), the progesterone receptor (Pgr) and signal transducer and activator of transcription 5a (Stat5a), factors that influence key mitogenic pathways that regulate normal mammary gland development.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Sustancias de Crecimiento/genética , Glándulas Mamarias Animales/crecimiento & desarrollo , Proteínas Nucleares/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Células Cultivadas , Estradiol/farmacología , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Sustancias de Crecimiento/metabolismo , Glándulas Mamarias Animales/efectos de los fármacos , Glándulas Mamarias Animales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfogénesis/efectos de los fármacos , Morfogénesis/genética , Morfogénesis/fisiología , Células 3T3 NIH , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína de Interacción con Receptores Nucleares 1 , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología
11.
FASEB J ; 26(10): 4198-209, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22750516

RESUMEN

Eutherian spermatozoa are dependent on the environment of the proximal epididymis to complete their maturation; however, no specific epididymal factors that mediate this process have so far been identified. Here, we show that targeted disruption of the novel gene Rnase10 encoding a secreted proximal epididymal protein in the mouse results in a binding defect in spermatozoa and their inability to pass through the uterotubal junction in the female. The failure to gain the site of fertilization in the knockout spermatozoa is associated with a gradual loss of ADAM3 and ADAM6 proteins during epididymal transit. In the distal epididymis, these spermatozoa appear to lack calcium-dependent associations with the immobilizing glutinous extracellular material and are released as single, vigorously motile cells that display no tendency for head-to-head agglutination and lack affinity to the oviductal epithelium. In sperm-egg binding assay, they are unable to establish a tenacious association with the zona pellucida, yet they are capable of fertilization. Furthermore, these sperm show accelerated capacitation resulting in an overall in vitro fertilizing ability superior to that of wild-type sperm. We conclude that the physiological role of sperm adhesiveness is in the mechanism of restricted sperm entry into the oviduct rather than in sperm-egg interaction.


Asunto(s)
Endorribonucleasas/metabolismo , Epidídimo/enzimología , Fertilidad/fisiología , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Animales , Endorribonucleasas/genética , Epidídimo/metabolismo , Femenino , Fertilidad/genética , Fertilización In Vitro , Immunoblotting , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Maduración del Esperma/genética , Maduración del Esperma/fisiología , Interacciones Espermatozoide-Óvulo/genética , Interacciones Espermatozoide-Óvulo/fisiología , Espermatozoides/metabolismo , Zona Pelúcida/metabolismo
12.
J Biol Rhythms ; 26(3): 187-99, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21628546

RESUMEN

Circadian rhythms are generated in central and peripheral tissues by an intracellular oscillating timing mechanism known as the circadian clock. Several lines of evidence show a strong and bidirectional interplay between metabolism and circadian rhythms. Receptor interacting protein 140 (RIP140) is a coregulator for nuclear receptors and other transcription factors that represses catabolic pathways in metabolic tissues. Although RIP140 functions as a corepressor for most nuclear receptors, mounting evidence points to RIP140 as a dual coregulator that can repress or activate different sets of genes. Here, we demonstrate that RIP140 mRNA and protein levels are under circadian regulation and identify RIP140 as a modulator of clock gene expression, suggesting that RIP140 can participate in a feedback mechanism affecting the circadian clock. We show that the absence of RIP140 disturbs the basal levels of BMAL1 and other clock genes, reducing the amplitude of their oscillations. In addition, we demonstrate that RIP140 is recruited to retinoid-related orphan receptor (ROR) binding sites on the BMAL1 promoter, directly interacts with RORα, and increases transcription from the BMAL1 promoter in a RORα-dependent manner. These results indicate that RIP140 is not only involved in metabolic control but also acts as a coactivator for RORα, influencing clock gene expression.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Relojes Circadianos/genética , Ritmo Circadiano , Regulación de la Expresión Génica , Proteínas Nucleares/metabolismo , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Factores de Transcripción ARNTL/metabolismo , Animales , Células Cultivadas , Fibroblastos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína de Interacción con Receptores Nucleares 1 , ARN Mensajero/metabolismo
13.
Neurosci Lett ; 425(2): 69-72, 2007 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-17822843

RESUMEN

Chronic exposure of rats to ethanol results in significant changes in pituitary hormone secretion. However, identification of the site(s) and mechanism of action of ethanol to induce these effects remains elusive. Free radical damage at the adenohypophyseal level may play a role in the decline in serum gonadotropin levels in ethanol-fed rats. Since 24-h changes in redox state occurred, we analyzed the 24-h changes in pituitary gene expression of the prooxidant enzymes nitric oxide synthase (NOS) 1 and 2, and of heme oxygenase-1 (HO-1) enzyme, and in plasma NO(2)(-) and NO(3)(-) (NO(x)) levels, in ethanol and control rats. Male rats, 35-day-old, received a liquid diet for 4 weeks. The ethanol-fed group received a similar diet to controls except for that maltose was isocalorically replaced by ethanol. Animals were killed at six time intervals during a 24-h cycle. Anterior pituitary mRNA levels encoding NOS1, NOS2 and HO-1 were measured by real-time PCR analysis. Plasma NO(x) concentration was determined by the Griess reaction. Ethanol feeding of prepubertal rats changed significantly the 24-h pattern of expression of NOS1, NOS2 and HO-1 in the adenohypophysis and augmented NOS2 and HO-1 mRNA levels. Peak values for the three enzymes in ethanol-fed rats occurred at the beginning of the scotophase (i.e., at 21:00 h). Ethanol feeding augmented mean values plasma NO(x) levels with a maximum at 13:00 h while in controls a biphasic pattern was observed, with peaks at 09:00 h and 17:00-21:00 h. One of the mechanisms by which ethanol augments oxidative damage in the adenohypophysis may include overproduction of nitric oxide and carbon monoxide.


Asunto(s)
Ritmo Circadiano/efectos de los fármacos , Etanol/efectos adversos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Hemo-Oxigenasa 1/genética , Óxido Nítrico Sintasa/genética , Adenohipófisis/efectos de los fármacos , Trastornos del Sistema Nervioso Inducidos por Alcohol/enzimología , Trastornos del Sistema Nervioso Inducidos por Alcohol/genética , Trastornos del Sistema Nervioso Inducidos por Alcohol/fisiopatología , Animales , Monóxido de Carbono/metabolismo , Depresores del Sistema Nervioso Central/efectos adversos , Ritmo Circadiano/genética , Radicales Libres/metabolismo , Regulación Enzimológica de la Expresión Génica/genética , Gonadotropinas Hipofisarias/metabolismo , Masculino , Óxido Nítrico Sintasa de Tipo I , Óxido Nítrico Sintasa de Tipo II/genética , Nitritos/sangre , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Enfermedades de la Hipófisis/inducido químicamente , Enfermedades de la Hipófisis/enzimología , Enfermedades de la Hipófisis/genética , Adenohipófisis/enzimología , Adenohipófisis/fisiopatología , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
14.
Toxicol Lett ; 172(3): 131-6, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17590544

RESUMEN

Chronic cadmium (Cd) administration affects the circadian release of pituitary hormones in rats. To assess whether Cd modifies expression of two major clock genes, period (Per) 1 and Per 2, in the hypothalamic-pituitary unit and to what extent the changes could be prevented by melatonin, rats were exposed to CdCl(2) (5ppm in drinking water) with or without melatonin (3 microg/mL drinking water) for 1 month and were killed at two time intervals, i.e. a the beginning of the rest span (09:00h) and at the middle of the activity span (01:00h). Hypothalamic and pituitary mRNA levels encoding Per 1 and Per 2 were measured by real-time PCR analysis. Cd treatment decreased expression of hypothalamic Per 1 gene at both time intervals, of hypothalamic Per 2 gene at 01:00h, and of adenohypophysial Per 1 and Per 2 genes at 09:00h. Melatonin administration counteracted most of the effects of Cd and augmented hypothalamic Per 2, and adenohypophysial Per 1 and Per 2 gene expression. The results indicate that Cd administered chronically in the drinking water to rats affected expression of clock genes in the hypothalamic-pituitary unit, an effect prevented by melatonin.


Asunto(s)
Cadmio/toxicidad , Proteínas de Ciclo Celular/genética , Hipotálamo/efectos de los fármacos , Melatonina/farmacología , Proteínas Nucleares/genética , Adenohipófisis/efectos de los fármacos , Animales , Antioxidantes/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Proteínas Circadianas Period , Adenohipófisis/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Wistar
15.
Nitric Oxide ; 16(2): 237-46, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16996755

RESUMEN

We previously showed that long-term exposure of anterior pituitary cells to nitric oxide (NO) induces apoptosis. The intracellular signals underlying this effect remained unclear. In this study, we searched for possible mechanisms involved in the early stages of the NO apoptotic cascade. Caspase 3 was activated by NO with no apparent disruption of mitochondrial membrane potential. NO caused a rapid increase of reactive oxygen species (ROS), and this increase seems to be dependent of mitochondrial electron transport chain. The antioxidant N-acetyl-cysteine avoided ROS increase, prevented the NO-induced caspase 3 activation, and reduced the NO apoptotic effect. Catalase was inactivated by NO, while glutathione peroxidase (GPx) activity and reduced glutathione (GSH) were not modified at first, but increased at later times of NO exposure. The increase of GSH level is important for the scavenging of the NO-induced ROS overproduction. Our results indicate that ROS have an essential role as a trigger of the NO apoptotic cascade in anterior pituitary cells. The permanent inhibition of catalase may strengthen the oxidative damage induced by NO. GPx activity and GSH level augment in response to the oxidative damage, though this increase seems not to be enough to rescue the cells from the NO effect.


Asunto(s)
Apoptosis/fisiología , Óxido Nítrico/fisiología , Adenohipófisis/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Antioxidantes/farmacología , Caspasa 3/metabolismo , Activación Enzimática , Femenino , Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , Potenciales de la Membrana , Mitocondrias/fisiología , Adenohipófisis/citología , Adenohipófisis/enzimología , Ratas , Ratas Wistar
16.
Toxicol Appl Pharmacol ; 218(1): 79-87, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17141818

RESUMEN

Hexavalent chromium (Cr VI) is a highly toxic metal and an environmental pollutant. Different studies indicate that Cr VI exposure adversely affects reproductive functions. This metal has been shown to affect several tissues and organs but Cr VI effects on pituitary gland have not been reported. Anterior pituitary hormones are central for the body homeostasis and have a fundamental role in reproductive physiology. The aim of this study was to evaluate the effect of Cr VI at the pituitary level both in vivo and in vitro. We showed that Cr VI accumulates in the pituitary and hypothalamus, and decreases serum prolactin levels in vivo but observed no effects on LH levels. In anterior pituitary cells in culture, the effect of Cr VI on hormone secretion followed the same differential pattern. Besides, lactotrophs were more sensitive to the toxicity of the metal. As a result of oxidative stress generation, Cr VI induced apoptosis evidenced by nuclear fragmentation and caspase 3 activation. Our results indicate that the anterior pituitary gland can be a target of Cr VI toxicity in vivo and in vitro, thus producing a negative impact on the hypothalamic-pituitary-gonadal axis and affecting the normal endocrine function.


Asunto(s)
Apoptosis/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Gonadotropinas Hipofisarias/metabolismo , Adenohipófisis/efectos de los fármacos , Dicromato de Potasio/toxicidad , Acetilcisteína/farmacología , Animales , Antioxidantes/farmacología , Caspasa 3/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cromo/toxicidad , Relación Dosis-Respuesta a Droga , Contaminantes Ambientales/metabolismo , Activación Enzimática/efectos de los fármacos , Gonadotropinas Hipofisarias/sangre , Hipotálamo/metabolismo , Hígado/metabolismo , Hormona Luteinizante/metabolismo , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Adenohipófisis/metabolismo , Adenohipófisis/patología , Dicromato de Potasio/metabolismo , Prolactina/metabolismo , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo
17.
J Pineal Res ; 41(3): 238-46, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16948784

RESUMEN

Cadmium (Cd) is widely used in industrial applications and is an important side contaminant of agricultural products. As an endocrine disruptor, Cd modifies pituitary hormone release. It has been shown that this metal causes oxidative stress in primary cultures of anterior pituitary cells. To examine whether Cd induces redox damage in the hypothalamic-pituitary axis in vivo and to evaluate the efficacy of the antioxidant molecule melatonin to prevent Cd activity, rats were exposed to Cd (5 p.p.m. in drinking water) with or without melatonin (3 microg/mL drinking water) for 1 month. In the anterior pituitary, Cd increased lipid peroxidation and mRNA levels for heme oxygenase-1 (HO-1) at both time intervals tested (09:00 and 01:00 hr, beginning of rest span and middle of activity span, respectively). Melatonin administration prevented the Cd-induced increase in both parameters. In the hypothalamus, Cd affected the levels of mRNA for HO-1 by decreasing it in the evening. Melatonin reduced hypothalamic HO-1 gene expression. Cd treatment augmented gene expression of nitric oxide synthase (NOS)1 and NOS2 in the pituitary whereas melatonin decreased it, impairing the activity of Cd. Exposure to Cd increased the levels of hypothalamic NOS1 mRNA at 09:00 hr and decreased the levels of NOS2 mRNA at 01:00 hr, with melatonin treatment preventing Cd effects. Cd treatment decreased plasma thyroid-stimulating hormone levels at both examined times, while melatonin reversed the effect of Cd at 09:00 hr and partially counteracted the effect at 01:00 hr. There were important variations between day and night in the expression of all the genes tested in both tissues. Melatonin treatment was effective reducing all examined effects of Cd, documenting its effectiveness to protect the rat hypothalamic-pituitary axis from the toxic metal effects.


Asunto(s)
Cadmio/farmacología , Expresión Génica/fisiología , Hipotálamo/metabolismo , Melatonina/fisiología , Adenohipófisis/metabolismo , Animales , Expresión Génica/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Masculino , Oxidación-Reducción/efectos de los fármacos , Adenohipófisis/efectos de los fármacos , Ratas , Ratas Wistar
18.
Endocrinology ; 147(9): 4311-8, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16740976

RESUMEN

Previous studies showed that 17 beta-estradiol (17 beta-E2) regulates the nitric oxide (NO)/soluble guanylyl cyclase (sGC)/cGMP pathway in many tissues. Evidence from our laboratory indicates that 17 beta-E2 disrupts the inhibitory effect of NO on prolactin release, decreasing sGC activity and affecting the cGMP pathway in anterior pituitary gland of adult ovariectomized and estrogenized rats. To ascertain the mechanisms by which 17 beta-E2 affects sGC activity, we investigated the in vivo and in vitro effects of 17 beta-E2 on sGC protein and mRNA expression in anterior pituitary gland from immature female rats. In the present work, we showed that 17 beta-E2 acute treatment exerted opposite effects on the two sGC subunits, increasing alpha1 and decreasing beta1 subunit protein and mRNA expression. This action on sGC protein expression was maximal 6-9 h after 17 beta-E2 administration. 17beta-E2 also caused the same effect on mRNA expression at earlier times. Concomitantly, 17 beta-E2 dramatically decreased sGC activity 6 and 9 h after injection. These effects were specific of 17 beta-E2, because they were not observed with the administration of other steroids such as progesterone and 17 alpha-estradiol. This inhibitory action of 17beta-E2 on sGC also required the activation of estrogen receptor (ER), because treatment with the pure ER antagonist ICI 182,780 completely blocked 17 beta-E2 action. 17 beta-E2 acute treatment caused the same effects on pituitary cells in culture. These results suggest that 17 beta-E2 exerts an acute inhibitory effect on sGC in anterior pituitary gland by down-regulating sGC beta 1 subunit and sGC activity in a specific, ER-dependent manner.


Asunto(s)
Estradiol/farmacología , Guanilato Ciclasa/genética , Óxido Nítrico/farmacología , Adenohipófisis/enzimología , Animales , GMP Cíclico/biosíntesis , Regulación hacia Abajo/efectos de los fármacos , Estradiol/análogos & derivados , Femenino , Fulvestrant , Expresión Génica/efectos de los fármacos , Guanilato Ciclasa/análisis , Guanilato Ciclasa/metabolismo , Isoenzimas/análisis , Isoenzimas/genética , Isoenzimas/metabolismo , ARN Mensajero/análisis , Ratas , Ratas Wistar , Receptores de Estrógenos/antagonistas & inhibidores , Receptores de Estrógenos/fisiología , Solubilidad
19.
Free Radic Biol Med ; 40(4): 679-88, 2006 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-16458199

RESUMEN

Cadmium (Cd2+) is a highly toxic metal that affects the endocrine system. We have previously shown that Cd2+ induces caspase-3 activation and apoptosis of anterior pituitary cells and that endogenous nitric oxide (NO) protects these cells from Cd2+. Here we investigate the mechanisms by which NO exerts this protective role. Cd2+ (25 microM) reduced the mitochondrial membrane potential (MMP) as measured by flow cytometry. Cd2+-induced apoptosis was mitochondrial dependent since cyclosporin A protected the cells from this metal. Inhibition of NO synthesis with 0.5 mM L-NAME increased the effect of Cd2+ on MMP, whereas the NO donor DETANONOate (0.1 mM) reduced it. Cd2+ increased the production of reactive oxygen species (ROS) as measured by flow cytometry. This effect was electron-transfer-chain-dependent since it was inhibited by rotenone. In fact, rotenone reduced the cytotoxic effect of the metal. The action of Cd2+ on mitochondrial integrity was ROS dependent. Trolox, an antioxidant, inhibited the effect of the metal on the MMP. Cd2+-induced increase in ROS generation was reduced by DETANONOate. There are discrepancies concerning the role of NO in Cd2+ toxicity. Here we show that NO reduces Cd2+ toxicity by protecting the mitochondria from oxidative stress in a system where NO plays a regulatory role.


Asunto(s)
Apoptosis/efectos de los fármacos , Cadmio/toxicidad , Mitocondrias/efectos de los fármacos , Óxido Nítrico/farmacología , Estrés Oxidativo/efectos de los fármacos , Adenohipófisis/efectos de los fármacos , Animales , Células Cultivadas , Ciclosporina/farmacología , Citometría de Flujo , Inmunosupresores/farmacología , Masculino , Potenciales de la Membrana/efectos de los fármacos , Adenohipófisis/citología , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo
20.
Free Radic Biol Med ; 37(9): 1463-71, 2004 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-15454286

RESUMEN

Cadmium (Cd2+) is a potent toxic metal for both plants and animals. Chronic exposure to low doses of Cd2+ results in damage to several organs. We have previously reported that Cd2+ induces apoptosis in anterior pituitary cells by a caspase- and oxidative stress-dependent mechanism. Nitric oxide (NO) synthesis is affected by Cd2+ in several systems. NO has been shown to be either cytoprotective or cytotoxic in many systems. The aim of this study was to evaluate the possible participation of NO in the cytotoxic effect of Cd2+ on rat anterior pituitary cells. Cell viability was evaluated by mitochondrial dehydrogenase activity assay and confirmed by microscopy, studying nuclear morphology. Here we show that DETA NONOate ((Z)-1-[2 (2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium-1,2-diolate), a long-term NO donor, at concentrations below 0.5 mM, reduces nuclear condensation and fragmentation and reverses the decrease in cellular activity induced by Cd2+. Cd2+, by itself, induced NO synthesis, and inhibition of this synthesis enhanced Cd2+ cytotoxicity. NO also prevented caspase-3 activation and lipidic peroxidation induced by Cd2+. The NO/cGMP pathway does not seem to be involved in the cytoprotective effect of NO. These results indicate that NO has a cytoprotective role in Cd2+ -induced apoptosis, suggesting that endogenous NO could have a physiological role in protecting anterior pituitary cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Arginina/farmacología , Cadmio/toxicidad , Óxido Nítrico/farmacología , Adenohipófisis/efectos de los fármacos , Animales , Masculino , Donantes de Óxido Nítrico/farmacología , Compuestos Nitrosos/farmacología , Adenohipófisis/citología , Adenohipófisis/patología , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...