Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 221
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 9(1): 4194, 2018 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-30305620

RESUMEN

Puberty is regulated by epigenetic mechanisms and is highly sensitive to metabolic and nutritional cues. However, the epigenetic pathways mediating the effects of nutrition and obesity on pubertal timing are unknown. Here, we identify Sirtuin 1 (SIRT1), a fuel-sensing deacetylase, as a molecule that restrains female puberty via epigenetic repression of the puberty-activating gene, Kiss1. SIRT1 is expressed in hypothalamic Kiss1 neurons and suppresses Kiss1 expression. SIRT1 interacts with the Polycomb silencing complex to decrease Kiss1 promoter activity. As puberty approaches, SIRT1 is evicted from the Kiss1 promoter facilitating a repressive-to-permissive switch in chromatin landscape. Early-onset overnutrition accelerates these changes, enhances Kiss1 expression and advances puberty. In contrast, undernutrition raises SIRT1 levels, protracts Kiss1 repression and delays puberty. This delay is mimicked by central pharmacological activation of SIRT1 or SIRT1 overexpression, achieved via transgenesis or virogenetic targeting to the ARC. Our results identify SIRT1-mediated inhibition of Kiss1 as key epigenetic mechanism by which nutritional cues and obesity influence mammalian puberty.


Asunto(s)
Epigénesis Genética , Kisspeptinas/genética , Fenómenos Fisiológicos de la Nutrición , Obesidad/metabolismo , Maduración Sexual , Sirtuina 1/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Cromatina/metabolismo , Femenino , Histonas/metabolismo , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Ratones Transgénicos , Modelos Biológicos , Neuronas/metabolismo , Estado Nutricional , Complejo Represivo Polycomb 2/metabolismo , Regiones Promotoras Genéticas , Ratas , Ratas Wistar , Factores de Tiempo
2.
Reprod Domest Anim ; 52 Suppl 2: 354-358, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27859771

RESUMEN

The intent of this contribution is to provide an update of the progress we have made towards developing a method/treatment to permanently sterilize cats. Our approach employs two complementary methodologies: RNA interference (RNAi) to silence genes involved in the central control of reproduction and a virus-based gene therapy system intended to deliver RNAi selectively to the hypothalamus (where these genes are expressed) via the systemic administration of modified viruses. We selected the hypothalamus because it contains neurons expressing Kiss1 and Tac3, two genes essential for reproduction and fertility. We chose the non-pathogenic adeno-associated virus (AAV) as a vector whose tropism could be modified to target the hypothalamus. The issues that must be overcome to utilize this vector as a delivery vehicle to induce sterility include modification of the wild-type AAV to target the hypothalamic region of the brain with a simultaneous reduction in targeting of peripheral tissues and non-hypothalamic brain regions, identification of RNAi targets that will effectively reduce the expression of Kiss1 and Tac3 without off-target effects, and determination if neutralizing antibodies to the AAV serotype of choice are present in cats. Successful resolution of these issues will pave the way for the development of a powerful tool to induce the permanent sterility in cats.


Asunto(s)
Gatos , Anticoncepción/veterinaria , Dependovirus , Silenciador del Gen , Vectores Genéticos , Hipotálamo , Animales , Anticoncepción/métodos , Expresión Génica/efectos de los fármacos , Ingeniería Genética/métodos , Ingeniería Genética/veterinaria , Infertilidad/etiología , Infertilidad/veterinaria , Kisspeptinas/antagonistas & inhibidores , Kisspeptinas/genética , Neuroquinina B/antagonistas & inhibidores , Neuroquinina B/genética , Interferencia de ARN
3.
Cell Death Dis ; 6: e1685, 2015 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-25766324

RESUMEN

Proliferation, differentiation and death of ovarian cells ensure orderly functioning of the female gonad during the reproductive phase, which ultimately ends with menopause in women. These processes are regulated by several mechanisms, including local signaling via neurotransmitters. Previous studies showed that ovarian non-neuronal endocrine cells produce acetylcholine (ACh), which likely acts as a trophic factor within the ovarian follicle and the corpus luteum via muscarinic ACh receptors. How its actions are restricted was unknown. We identified enzymatically active acetylcholinesterase (AChE) in human ovarian follicular fluid as a product of human granulosa cells. AChE breaks down ACh and thereby attenuates its trophic functions. Blockage of AChE by huperzine A increased the trophic actions as seen in granulosa cells studies. Among ovarian AChE variants, the readthrough isoform AChE-R was identified, which has further, non-enzymatic roles. AChE-R was found in follicular fluid, granulosa and theca cells, as well as luteal cells, implying that such functions occur in vivo. A synthetic AChE-R peptide (ARP) was used to explore such actions and induced in primary, cultured human granulosa cells a caspase-independent form of cell death with a distinct balloon-like morphology and the release of lactate dehydrogenase. The RIPK1 inhibitor necrostatin-1 and the MLKL-blocker necrosulfonamide significantly reduced this form of cell death. Thus a novel non-enzymatic function of AChE-R is to stimulate RIPK1/MLKL-dependent regulated necrosis (necroptosis). The latter complements a cholinergic system in the ovary, which determines life and death of ovarian cells. Necroptosis likely occurs in the primate ovary, as granulosa and luteal cells were immunopositive for phospho-MLKL, and hence necroptosis may contribute to follicular atresia and luteolysis. The results suggest that interference with the enzymatic activities of AChE and/or interference with necroptosis may be novel approaches to influence ovarian functions.


Asunto(s)
Acetilcolinesterasa/biosíntesis , Células de la Granulosa/enzimología , Folículo Ovárico/metabolismo , Proteínas Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Acetilcolina/metabolismo , Acetilcolinesterasa/metabolismo , Acrilamidas/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Diferenciación Celular/genética , Femenino , Células de la Granulosa/efectos de los fármacos , Humanos , Imidazoles/administración & dosificación , Indoles/administración & dosificación , Folículo Ovárico/crecimiento & desarrollo , Cultivo Primario de Células , Proteínas Quinasas/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Sulfonamidas/administración & dosificación
4.
Endocrinology ; 154(2): 942-55, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23291449

RESUMEN

Lin28 and Lin28b are related RNA-binding proteins that inhibit the maturation of miRNAs of the let-7 family and participate in the control of cellular stemness and early embryonic development. Considerable interest has arisen recently concerning other physiological roles of the Lin28/let-7 axis, including its potential involvement in the control of puberty, as suggested by genome-wide association studies and functional genomics. We report herein the expression profiles of Lin28 and let-7 members in the rat hypothalamus during postnatal maturation and in selected models of altered puberty. The expression patterns of c-Myc (upstream positive regulator of Lin28), mir-145 (negative regulator of c-Myc), and mir-132 and mir-9 (putative miRNA repressors of Lin28, predicted by bioinformatic algorithms) were also explored. In male and female rats, Lin28, Lin28b, and c-Myc mRNAs displayed very high hypothalamic expression during the neonatal period, markedly decreased during the infantile-to-juvenile transition and reached minimal levels before/around puberty. A similar puberty-related decline was observed for Lin28b in monkey hypothalamus but not in the rat cortex, suggesting species conservation and tissue specificity. Conversely, let-7a, let-7b, mir-132, and mir-145, but not mir-9, showed opposite expression profiles. Perturbation of brain sex differentiation and puberty, by neonatal treatment with estrogen or androgen, altered the expression ratios of Lin28/let-7 at the time of puberty. Changes in the c-Myc/Lin28b/let-7 pathway were also detected in models of delayed puberty linked to early photoperiod manipulation and, to a lesser extent, postnatal underfeeding or chronic subnutrition. Altogether, our data are the first to document dramatic changes in the expression of the Lin28/let-7 axis in the rat hypothalamus during the postnatal maturation and after different manipulations that disturb puberty, thus suggesting the potential involvement of developmental changes in hypothalamic Lin28/let-7 expression in the mechanisms permitting/leading to puberty onset.


Asunto(s)
Envejecimiento/genética , Encéfalo/crecimiento & desarrollo , MicroARNs/metabolismo , Proteínas de Unión al ARN/biosíntesis , Animales , Células Madre Embrionarias/citología , Femenino , Hipotálamo/crecimiento & desarrollo , Hipotálamo/metabolismo , Masculino , MicroARNs/biosíntesis , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Pubertad/efectos de los fármacos , Ratas , Ratas Wistar , Distribución Tisular
5.
Hum Reprod ; 27(11): 3249-58, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22888166

RESUMEN

STUDY QUESTION: Is decorin (DCN), a putative modulator of growth factor (GF) signaling, expressed in the primate ovary and does it play a role in ovarian biology? SUMMARY ANSWER: DCN expression in the theca, the corpus luteum (CL), its presence in the follicular fluid (FF) and its actions revealed in human IVF-derived granulosa cells (GCs), suggest that it plays multiple roles in the ovary including folliculogenesis, ovulation and survival of the CL. WHAT IS KNOWN ALREADY: DCN is a secreted proteoglycan, which has a structural role in the extracellular matrix (ECM) and also interferes with the signaling of multiple GF/GF receptors (GFRs). However, DCN expression and action in the primate ovary has yet to be determined. STUDY DESIGN, SIZE, DURATION: Archival human and monkey ovarian samples were analyzed. Studies were conducted using FF and GC samples collected from IVF patients. PARTICIPANTS/MATERIALS, SETTING, METHODS: Immunohistochemistry, western blotting, RT-PCR, quantitative RT-PCR (qPCR) and enzyme-linked immunosorbent assay (ELISA) studies were complemented by cellular studies, including the measurements of intracellular Ca²âº, reactive oxygen species (ROS), epidermal GF receptor (EGFR) phosphorylation by DCN and caspase activity. MAIN RESULTS AND THE ROLE OF CHANCE: Immunohistochemistry revealed strong DCN staining in the connective tissue and follicular thecal compartments, but not in GCs of pre-antral and antral follicles. Pre-ovulatory follicles could not be studied, but DCN was associated with connective tissue of CL samples and the cytoplasm of luteal cells. DCN expression in monkey CL doubled (P < 0.05) towards the end of the luteal lifespan. DCN was found in human FF obtained from IVF patients (mean: 12.9 ng/ml; n = 20) as determined by ELISA. DCN mRNA and/or protein were detected in freshly isolated and cultured, luteinized human GCs. In the latter, exogenous human recombinant DCN increased intracellular Ca²âº levels and induced the production of ROS in a concentration-dependent manner. DCN, like epidermal GF, phosphorylated EGFR significantly (P < 0.05) and reduced the activity of caspase 3/7 in cultured GCs. The data indicate the expression of DCN in the theca of growing follicles, in FF of ovulatory follicles and in the CL. Therefore, DCN may exert paracrine actions via GF/GFR systems in multiple ovarian compartments. LIMITATIONS, REASONS FOR CAUTION: Functional studies were performed in cultures of human luteinized GCs, which are an apt model but may not fully mirror the pre-ovulatory GC compartment or the CL. Other human ovarian cells, including the thecal cells, were not available. WIDER IMPLICATIONS OF THE FINDINGS: In accordance with its evolving roles in other organs, ovarian DCN is an ECM-associated component, which acts as a multifunctional regulator of GF signaling in the primate ovary. DCN may thus be involved in folliculogenesis, ovulation and the regulation of the CL survival in primates. STUDY FUNDING/COMPETING INTEREST(S): This study was supported by Deutsche Forschungsgemeinschaft (DFG) MA1080/17-3 and in part DFG MA1080/21-1 (to AM), NIH grants HD24870 (S.R.O. and R.L.S.), the Eunice Kennedy Shriver NICHD/NIH through cooperative agreement HD18185 as part of the Specialized Cooperative Centers Program in Reproduction and Infertility Research (S.R.O.) and 8P51OD011092-53 for the operation of the Oregon National Primate Research Center (G.A.D., J.D.H., S.R.O. and R.L.S).


Asunto(s)
Decorina/metabolismo , Matriz Extracelular/metabolismo , Fase Luteínica/metabolismo , Oogénesis , Ovario/metabolismo , Ovulación/metabolismo , Adulto , Animales , Células Cultivadas , Cuerpo Lúteo/citología , Cuerpo Lúteo/metabolismo , Decorina/genética , Receptores ErbB/metabolismo , Femenino , Líquido Folicular/metabolismo , Regulación de la Expresión Génica , Células de la Granulosa/citología , Células de la Granulosa/metabolismo , Humanos , Macaca mulatta , Ovario/citología , Fosforilación , Procesamiento Proteico-Postraduccional , ARN Mensajero/metabolismo , Células Tecales/citología , Células Tecales/metabolismo
6.
Reprod Domest Anim ; 47 Suppl 4: 228-32, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22827375

RESUMEN

A non-surgical method to induce sterility would be a useful tool to control feral populations of animals. Our laboratories have experience with approaches aimed at targeting brain cells in vivo with vehicles that deliver a payload of either inhibitory RNAs or genes intended to correct cellular dysfunction. A combination/modification of these methods may provide a useful framework for the design of approaches that can be used to sterilize cats and dogs. For this approach to succeed, it has to meet several conditions: it needs to target a gene essential for fertility. It must involve a method that can selectively silence the gene of interest. It also needs to deliver the silencing agent via a minimally invasive method. Finally, the silencing effect needs to be sustained for many years, so that expansion of the targeted population can be effectively prevented. In this article, we discuss this subject and provide a succinct account of our previous experience with: (i) molecular reagents able to disrupt reproductive cyclicity when delivered to regions of the brain involved in the control of reproduction and (ii) molecular reagents able to ameliorate neuronal disease when delivered systemically using a novel approach of gene therapy.


Asunto(s)
Interferencia de ARN/fisiología , Esterilización Reproductiva/veterinaria , Adenoviridae , Animales , Gatos , Perros , Femenino , Fertilidad/fisiología , Vectores Genéticos , Hipotálamo/fisiología , Infertilidad Femenina , Masculino , MicroARNs , Regulación de la Población , Primates , Ratas , Esterilización Reproductiva/métodos
7.
J Neuroendocrinol ; 24(6): 916-29, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22356123

RESUMEN

Thyroid transcription factor 1 (TTF1), a member of the Nkx family of transcription factors required for basal forebrain morphogenesis, functions in the postnatal hypothalamus as a transcriptional regulator of genes encoding neuromodulators and hypophysiotrophic peptides. One of these peptides is gonadotrophin-releasing hormone (GnRH). In the present study, we show that Ttf1 mRNA abundance varies in a diurnal and melatonin-dependent fashion in the preoptic area of the rat, with maximal Ttf1 expression attained during the dark phase of the light/dark cycle, preceding the nocturnal peak in GnRH mRNA content. GnRH promoter activity oscillates in a circadian manner in GT1-7 cells, and this pattern is enhanced by TTF1 and blunted by small interfering RNA-mediated Ttf1 gene silencing. TTF1 transactivates GnRH transcription by binding to two sites in the GnRH promoter. Rat GnRH neurones in situ contain key proteins components of the positive (BMAL1, CLOCK) and negative (PER1) limbs of the circadian oscillator, and these proteins repress Ttf1 promoter activity in vitro. By contrast, Ttf1 transcription is activated by CRY1, a clock component required for circadian rhythmicity. In turn, TTF1 represses transcription of Rev-erbα, a heme receptor that controls circadian transcription within the positive limb of the circadian oscillator. These findings suggest that TTF1 is a component of the molecular machinery controlling circadian oscillations in GnRH gene transcription.


Asunto(s)
Ritmo Circadiano/genética , Regulación de la Expresión Génica/genética , Hormona Liberadora de Gonadotropina/fisiología , Proteínas de Homeodominio/fisiología , Proteínas Nucleares/fisiología , Transactivadores/fisiología , Factores de Transcripción/fisiología , Animales , Ritmo Circadiano/fisiología , Hormona Liberadora de Gonadotropina/biosíntesis , Hormona Liberadora de Gonadotropina/genética , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Ratas , Ratas Sprague-Dawley , Factor Nuclear Tiroideo 1 , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
8.
Reprod Domest Anim ; 47 Suppl 6: 381-6, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23279544

RESUMEN

Population control of feral animals is often difficult, as it can be dangerous for the animals, labour intensive and expensive. Therefore, a useful tool for control of animal populations would be a non-surgical method to induce sterility. Our laboratories utilize methods aimed at targeting brain cells in vivo with vehicles that deliver a payload of either inhibitory RNAs or genes intended to correct cellular dysfunction. A useful framework for design of a new approach will be the combination of these methods with the intended goal to produce a technique that can be used to non-invasively sterilize cats and dogs. For this approach to succeed, it has to meet several conditions: the target gene must be essential for fertility; the method must include a mechanism to effectively and specifically silence the gene of interest; the method of delivering the silencing agent must be minimally invasive, and finally, the silencing effect must be sustained for the lifespan of the target species, so that expansion of the population can be effectively prevented. In this article, we discuss our work to develop gene silencing technology to induce sterility; we will use examples of our previous studies demonstrating that this approach is viable. These studies include (i) the use of viral vectors able to disrupt reproductive cyclicity when delivered to the regions of the brain involved in the control of reproduction and (ii) experiments with viral vectors that are able to ameliorate neuronal disease when delivered systemically using a novel approach of gene therapy.


Asunto(s)
Gatos , Anticoncepción/veterinaria , Perros , Silenciador del Gen/fisiología , Esterilización Reproductiva/veterinaria , Animales , Anticoncepción/métodos , Femenino , Fertilidad/fisiología , Hipotálamo/fisiología , Masculino , MicroARNs , Regulación de la Población , Esterilización Reproductiva/métodos
9.
Hum Reprod ; 25(4): 969-76, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20097922

RESUMEN

BACKGROUND: Oxytocin (OT) is produced by granulosa cells (GCs) of pre-ovulatory ovarian follicles and the corpus luteum (CL) in some mammalian species. Actions of OT in the ovary have been linked to luteinization, steroidogenesis and luteolysis. Human IVF-derived (h)GCs possess a functional OT receptor (OTR), linked to elevation of intracellular Ca(2+), but molecular identity of the receptor for OT in human granulosa cells (hGCs) and down-stream consequences are not known. METHODS AND RESULTS: RT-PCR, sequencing and immunocytochemistry identified the genuine OTR in hGCs. OT (10 nM-10 microM) induced elevations of intracellular Ca(2+) levels (Fluo-4 measurements), which were blocked by tocinoic acid (TA; 50 microM, a selective OTR-antagonist). Down-stream effects of OTR-activation include a concentration dependent decrease in cell viability/metabolism, manifested by reduced ATP-levels, increased caspase3/7-activity (P < 0.05) and electron microscopical signs of cellular regression. TA blocked all of these changes. Immunoreactive OTR was found in the CL and GCs of large and, surprisingly, also small pre-antral follicles of the human ovary. Immunoreactive OTR in the rhesus monkey ovary was detected in primordial and growing primary follicles in the infantile ovary and in follicles at all stages of development in the adult ovary, as well as the CL: these results were corroborated by RT-PCR analysis of GCs excised by laser capture microdissection. CONCLUSIONS: Our study identifies genuine OTRs in human and rhesus monkey GCs. Activation by high levels of OT leads to cellular regression in hGCs. As GCs of small follicles also express OTRs, OT may have as yet unknown functions in follicular development.


Asunto(s)
Apoptosis/genética , Apoptosis/fisiología , Células de la Granulosa/citología , Células de la Granulosa/metabolismo , Macaca mulatta/genética , Macaca mulatta/metabolismo , Ovario/citología , Ovario/metabolismo , Receptores de Oxitocina/genética , Receptores de Oxitocina/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Secuencia de Bases , Señalización del Calcio , Cuerpo Lúteo/citología , Cuerpo Lúteo/metabolismo , Cartilla de ADN/genética , Femenino , Humanos , Inmunohistoquímica , Técnicas In Vitro , Microscopía Electrónica de Transmisión , Oxitocina/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Especificidad de la Especie
10.
J Neuroendocrinol ; 21(2): 108-22, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19187398

RESUMEN

The excitatory tone to gonadotrophin-releasing hormone (GnRH) neurones is a critical component underlying the pubertal increase in GnRH secretion. However, the homeostatic mechanisms modulating the response of GnRH neurones to excitatory inputs remain poorly understood. A basic mechanism of neuronal homeostasis is the Na(+),K(+)-ATPase-dependent restoration of Na(+) and K(+) transmembrane gradients after neuronal excitation. This activity is reduced in a mouse model of Rett syndrome (RTT), a neurodevelopmental disorder in which expression of FXYD1, a modulator of Na(+),K(+)-ATPase activity, is increased. We now report that the initiation, but not the completion of puberty, is advanced in girls with RTT, and that, in rodents, FXYD1 may contribute to the neuroendocrine regulation of female puberty by modulating GnRH neuronal excitability. Fxyd1 mRNA abundance reaches maximal levels in the female rat hypothalamus by the fourth postnatal week of life (i.e., around the time when the mode of GnRH secretion acquires an adult pattern of release). Although Fxyd1 mRNA expression is low in the hypothalamus, approximately 50% of GnRH neurones contain Fxyd1 transcripts. Whole-cell patch recording of GnRH-EGFP neurones revealed that the neurones of Fxyd1-null female mice respond to somatic current injections with a lower number of action potentials than wild-type cells. Both the age at vaginal opening and at first oestrous were delayed in Fxyd1(-/-) mice, but adult reproductive capacity was normal. These results suggest that FXYD1 contributes to facilitating the advent of puberty by maintaining GnRH neuronal excitability to incoming transsynaptic stimulatory inputs.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Proteínas de la Membrana/metabolismo , Neuronas/fisiología , Fosfoproteínas/metabolismo , Desarrollo Sexual/fisiología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Potenciales de Acción/fisiología , Adolescente , Animales , Niño , Preescolar , Femenino , Humanos , Hipotálamo/citología , Hipotálamo/metabolismo , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Técnicas de Placa-Clamp , Fosfoproteínas/genética , Pubertad/fisiología , Ratas
11.
J Neuroendocrinol ; 20(6): 732-42, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18601696

RESUMEN

A wealth of information now exists showing that glial cells are actively involved in the cell-cell communication process generating and disseminating information within the central nervous system. In the hypothalamus, two types of glial cells, astrocytes and ependymal cells lining the latero-ventral portion of the third ventricle (known as tanycytes), regulate the secretory activity of neuroendocrine neurones. This function, initially described for astrocytes apposing magnocellular neurones, has been more recently characterised for neurones secreting gonadotrophin hormone-releasing hormone (GnRH). The available evidence suggests that glial cells of the median eminence regulate GnRH secretion via two related mechanisms. One involves the production of growth factors acting via receptors with tyrosine kinase activity. The other involves plastic rearrangements of glia-GnRH neurone adhesiveness. GnRH axons reach the median eminence, at least in part, directed by basic fibroblast growth factor. Their secretory activity is facilitated by insulin-like growth factor 1 and members of the epidermal growth factor family. A structural complement to these soluble molecules is provided by at least three cell-cell adhesion systems endowed with signalling capabilities. One of them uses the neuronal cell adhesion molecule (NCAM), another employs the synaptic cell adhesion molecule (SynCAM), and the third one consists of neuronal contactin interacting with glial receptor-like protein tyrosine phosphatase-beta. It is envisioned that, within the median eminence, soluble factors and adhesion molecules work coordinately to control delivery of GnRH to the portal vasculature.


Asunto(s)
Comunicación Celular/fisiología , Hormona Liberadora de Gonadotropina/metabolismo , Eminencia Media/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Animales , Moléculas de Adhesión Celular Neuronal/metabolismo , Forma de la Célula , Factor de Crecimiento Epidérmico/metabolismo , Ácido Glutámico/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Eminencia Media/citología , Neuroglía/citología , Plasticidad Neuronal/fisiología , Neuronas/citología , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/metabolismo
12.
J Neuroendocrinol ; 19(11): 847-59, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17927663

RESUMEN

Although it is well established that gonadotrophin-releasing hormone (GnRH) neurones and astrocytes maintain an intimate contact throughout development and adult life, the cell-surface molecules that may contribute to this adhesiveness remain largely unknown. In the peripheral nervous system, the glycosylphosphatidyl inositol (GPI)-anchored protein contactin is a cell-surface neuronal protein required for axonal-glial adhesiveness. A glial transmembrane protein recognised by neuronal contactin is receptor-like protein tyrosine phosphatase beta (RPTP beta), a phosphatase with structural similarities to cell adhesion molecules. In the present study, we show that contactin, and its preferred in cis partner Caspr1, are expressed in GnRH neurones. We also show that the RPTP beta mRNA predominantly expressed in hypothalamic astrocytes encodes an RPTP beta isoform (short RPTP beta) that uses its carbonic anhydrase (CAH) extracellular subdomain to interact with neuronal contactin. Immunoreactive contactin is most abundant in GnRH nerve terminals projecting to both the organum vasculosum of the lamina terminalis and median eminence, implying GnRH axons as an important site of contactin-dependent cell adhesiveness. GT1-7 immortalised GnRH neurones adhere to the CAH domain of RPTPbeta, and this adhesiveness is blocked when contactin GPI anchoring is disrupted or contactin binding capacity is immunoneutralised, suggesting that astrocytic RPTP beta interacts with neuronal contactin to mediate glial-GnRH neurone adhesiveness. Because the abundance of short RPTP beta mRNA increases in the female mouse hypothalamus (but not in the cerebral cortex) before puberty, it appears that an increased interaction between GnRH axons and astrocytes mediated by RPTP beta-contactin is a dynamic mechanism of neurone-glia communication during female sexual development.


Asunto(s)
Astrocitos/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Adhesión Celular/fisiología , Comunicación Celular/fisiología , Neuronas/metabolismo , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/metabolismo , Animales , Astrocitos/citología , Moléculas de Adhesión Celular Neuronal/genética , Células Cultivadas , Contactinas , Femenino , Hormona Liberadora de Gonadotropina/genética , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/citología , Hipotálamo/fisiología , Ratones , Neuronas/citología , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
13.
Endocrinology ; 147(8): 3789-96, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16728490

RESUMEN

Mammalian ovaries contain sympathetic neurons expressing the low affinity neurotropin receptor (p75NTR). To date neither the role these neurons might play in ovarian physiology nor their embryological origin is known. Immunohistochemistry was used to detect postnatal changes in distribution and number of both p75NTR-positive and tyrosine hydroxylase-positive neurons in rhesus monkey ovaries. Pig fetuses were used to map the pathway of ovarian neuronal migration during embryonic development. Antiserum to p75NTR revealed the presence of isolated neurons and neurons clustered into ganglia in 2-month-old monkey ovaries. After 8 months, the neurons exhibited well-developed processes, and other than being more extensively interlaced, the localization and morphology did not change after 2 yr of age. Total number of p75NTR-positive neurons present decreased gradually between 2 months and 12 yr of age and declined markedly with reproductive aging. Conversely, the subpopulation of neurons immunoreactive to anti-tyrosine hydroxylase increased significantly at puberty and then declined with the loss of reproductive capacity. By d 21 of fetal life in the pig, p75NTR neurons had migrated medially from the neural crest to form the paraaortic autonomic ganglia. Some neurons migrated ventrally from the ganglia and then continued ventrolaterally to enter the genital ridge. By d 27, neurons had entered the developing ovary, and by d 35, the migration was complete with neurons demonstrating immunoreactivity to NeuN, a neuron-specific marker. Results demonstrate that p75NTR-expressing ovarian neurons originate from the neural crest and that a catecholaminergic subset is associated with pubertal maturation of the ovary and subsequent reproductive function.


Asunto(s)
Ganglios Simpáticos/citología , Ganglios Simpáticos/crecimiento & desarrollo , Neuronas/citología , Ovario/crecimiento & desarrollo , Ovario/inervación , Factores de Edad , Envejecimiento/fisiología , Animales , Recuento de Células , Femenino , Ganglios Simpáticos/embriología , Macaca mulatta , Mamíferos , Cresta Neural/citología , Cresta Neural/embriología , Neuronas/metabolismo , Ovario/embriología , Receptor de Factor de Crecimiento Nervioso/metabolismo , Maduración Sexual/fisiología , Porcinos , Tirosina 3-Monooxigenasa/metabolismo
14.
J Clin Endocrinol Metab ; 91(6): 2396-403, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16537688

RESUMEN

CONTEXT: Previous studies showed that nerve growth factor (NGF) induces the expression of functional FSH receptors (FSHR) in preantral follicles of the developing rat ovary. OBJECTIVE: The objective of this study was to determine whether NGF can affect granulosa cell (GC) function in human periovulatory follicles using intact human ovaries and isolated human GCs. PATIENTS AND INTERVENTIONS: Human GCs were obtained from in vitro fertilization patients and normal ovaries from women with elective pelvic surgery for nonovarian indications. RESULTS: In normal ovaries, NGF and trkA (NGF's high-affinity receptor) were detected by immunohistochemistry in GCs of preantral and antral follicles. NGF and trkA are also present in thecal cells of antral follicles. Both freshly collected and cultured GCs contained immunoreactive NGF and trkA in addition to their respective mRNAs. Human GCs respond to NGF with increased estradiol (E(2)) secretion and a reduction in progesterone output. Exposure of human GCs to NGF increased FSHR mRNA content within 18 h of treatment, and this effect was blocked by the trk tyrosine kinase blocker K-252a. Also, cells preexposed to NGF released significantly more E(2) in response to hFSH than cells not pretreated with the neurotropin, showing that the NGF-induced increase in FSHR gene expression results in the formation of functional FSHRs. CONCLUSIONS: These results suggest that one of the functions of NGF in the preovulatory human ovary is to increase the secretion of E(2) while preventing early luteinization via an inhibitory effect on progesterone secretion. NGF stimulates E(2) secretion both directly and by increasing the formation of FSHRs.


Asunto(s)
Estradiol/metabolismo , Células de la Granulosa/metabolismo , Factor de Crecimiento Nervioso/farmacología , Receptor trkA/fisiología , Receptores de HFE/biosíntesis , Femenino , Humanos , Progesterona/metabolismo , ARN Mensajero/análisis , Receptores de HFE/genética
15.
J Neuroendocrinol ; 17(4): 208-19, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15842232

RESUMEN

Transsynaptic and glial-neuronal communication are important components of the mechanism underlying the pubertal activation of luteinizing hormone-releasing hormone (LHRH) secretion. The molecules required for the architectural organization of these cell-cell interactions have not been identified. We now show that the hypothalamus of the prepubertal female rhesus monkey expresses a multiplicity of genes encoding three families of adhesion/signalling proteins involved in the structural definition of both neurone-to-neurone and bi-directional neurone-glia communication. These include the neurexin/neuroligin (NRX/NRL) and protocadherin-alpha (PCDHalpha) families of synaptic specifiers/adhesion molecules, and key components of the contactin-dependent neuronal-glial adhesiveness complex, including contactin/F3 itself, the contactin-associated protein-1 (CASPR1), and the glial receptor protein tyrosine phosphatase beta. Prominently expressed among members of the NRX family is the neurexin isoform involved in the specification of glutamatergic synapses. Although NRXs, PCDHalphas and CASPR1 transcripts are mostly detected in neurones, the topography of expression appears different. NRX1 mRNA-containing neurones are scattered throughout the hypothalamus, PCDHalpha mRNA transcripts appear more abundant in neurones of the arcuate nucleus and periventricular region, and neurones positive for CASPR1 mRNA exhibit a particularly striking distribution pattern that delineates the hypothalamus. Examination of LHRH neurones, using the LHRH-secreting cell line GT1-7, showed that these cells contain transcripts encoding NRXs and one of their ligands (NRL1), at least one PCDHalpha (CNR-8/PCDHalpha10), and the CASPR1/contactin complex. The results indicate that the prepubertal female monkey hypothalamus contains a plethora of adhesion/signalling molecules with different but complementary functions, and that an LHRH neuronal cell line expresses key components of this structural complex. The presence of such cell-cell communication machinery in the neuroendocrine brain suggests an integrated participation of their individual components in the central control of female sexual development.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Comunicación Celular/fisiología , Hipotálamo/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Maduración Sexual/fisiología , Animales , Cadherinas/genética , Cadherinas/metabolismo , Moléculas de Adhesión Celular/genética , Comunicación Celular/genética , Femenino , Perfilación de la Expresión Génica , Glicoproteínas/genética , Glicoproteínas/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Hipotálamo/citología , Macaca mulatta , Proteínas de la Membrana/genética , Familia de Multigenes/genética , Familia de Multigenes/fisiología , Proteínas del Tejido Nervioso/genética , Neuroglía/metabolismo , Neuronas/metabolismo , Neuropéptidos/genética , Neuropéptidos/metabolismo , Sistemas Neurosecretores/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Maduración Sexual/genética
16.
Cell Tissue Res ; 306(2): 231-7, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11702234

RESUMEN

Mammalian ovarian function is regulated by both hormonal inputs and direct neural influences. Recent studies have shown that, in addition to the extrinsic innervation, the ovaries of nonhuman primates and a strain of rats contain a discrete population of intrinsic neurons. In the present study, we used histological and immunohistochemical approaches to identify the presence of neuronal cell bodies in the fetal and neonatal human ovary. Neurons containing neurofilament immunoreactivity were detected in the hilum and medulla of the ovary at all ages studied, ranging from 24 weeks of gestation to 10 months of postnatal age. Most of them coexpressed the low affinity neurotrophin receptor (p75NTR), and some were catecholaminergic, as determined by their content of immunoreactive tyrosine hydroxylase (TH), the rate-limiting enzyme in catecholamine biosynthesis. The presence of intrinsic neurons in the human ovary, similar to those previously found in other species, indicates that they may be engaged in regulating common, phylogenetically conserved, ovarian functions. It also raises the possibility that their dysfunction may contribute to the manifestation of particular ovarian pathologies.


Asunto(s)
Neuronas/citología , Ovario/inervación , Animales , Biomarcadores , Tamaño de la Célula , Femenino , Feto , Humanos , Inmunohistoquímica/métodos , Proteínas de Neurofilamentos/análisis , Neuronas/química , Ovario/química , Ovario/citología , Ovario/embriología , Receptor de Factor de Crecimiento Nervioso , Receptores de Factor de Crecimiento Nervioso/análisis , Tirosina 3-Monooxigenasa/análisis
17.
Endocrinology ; 142(5): 2078-86, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11316775

RESUMEN

Nerve growth factor (NGF) epitomizes a family of proteins known as the neurotrophins (NTs), which are required for the survival and differentiation of neurons within both the central and peripheral nervous system. Synthesis of NGF in tissues innervated by the peripheral nervous system is consistent with its function as a target-derived trophic factor. However, the presence of low- and high-affinity NGF receptors in the gonads suggests another function for the NTs within the reproductive endocrine system. We now report that NGF is required for the growth of primordial ovarian follicles, a process known to occur independently of pituitary gonadotropins. Both the NT receptor p75(NTR) and the NGF tyrosine kinase receptor trkA were found to be expressed in the ovaries of infantile normal mice and mice carrying a null mutation of the NGF gene. The ovaries from homozygote NGF-null (-/-) mutant animals, analyzed after completion of ovarian histogenesis, exhibited a markedly reduced population of primary and secondary follicles in the presence of normal serum gonadotropin levels, and an increased number of oocytes that failed to be incorporated into a follicular structure. Assessment of mitogenic activity using two complementary proliferation markers revealed a conspicuous reduction in somatic cell proliferation in the ovaries of NGF-deficient mice. These results suggest that the delay in follicular growth observed in NGF(-/-) mice may be related to the loss of a proliferative signal provided by NGF to the nonneural endocrine component of the ovary.


Asunto(s)
Factor de Crecimiento Nervioso/fisiología , Folículo Ovárico/fisiología , Receptor trkA/genética , Animales , División Celular , Femenino , Ratones , Ratones Endogámicos C57BL , Factor de Crecimiento Nervioso/genética , Ovario/patología , Antígeno Nuclear de Célula en Proliferación/análisis , Receptor de Factor de Crecimiento Nervioso , Receptor trkA/análisis , Receptores de Factor de Crecimiento Nervioso/análisis
18.
Endocrinology ; 142(5): 2102-14, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11316778

RESUMEN

Gamma amino butyric acid (GABA) is the main inhibitory neurotransmitter controlling LH-releasing hormone (LHRH) secretion in the mammalian hypothalamus. Whether alterations in GABA homeostasis within discrete regions of the neuroendocrine brain known to be targets of GABA action, such as the median eminence, can disrupt the ability of the LHRH releasing system to maintain reproductive cyclicity is not known but amenable to experimental scrutiny. The present experiments were undertaken to examine this issue. Immortalized BAS-8.1 astroglial cells were genetically modified by infection with a regulatable retroviral vector to express the gene encoding the GABA synthesizing enzyme glutamic acid decarboxylase-67 (GAD-67) under the control of a tetracycline (tet) controlled gene expression system. In this system, expression of the gene of interest is repressed by tet and activated in the absence of the antibiotic. BAS-8.1 cells carrying this regulatory cassette, and cultured in the absence of tet ("GAD on"), expressed abundant levels of GAD-67 messenger RNA and GAD enzymatic activity, and released GABA when challenged with glutamate. All of these responses were inhibited within 24 h of exposure to tet ("GAD off"). Grafting "GAD on" cells into the median eminence of late juvenile female rats, near LHRH nerve terminals, did not affect the age at vaginal opening, but greatly disrupted subsequent estrous cyclicity. These animals exhibiting long periods of persistent estrus, interrupted by occasional days in proestrus and diestrus, suggesting the occurrence of irregular ovulatory episodes. Administration of the tetracycline analog doxycycline (DOXY) in the drinking water inhibited GAD-67synthesis and restored estrous cyclicity to a pattern indistinguishable from that of control rats grafted with native BAS-8.1 cells. Animals carrying "GAD on" cells showed a small increase in serum LH and estradiol levels, and a marked elevation in serum androstenedione, all of which were obliterated by turning GAD-67 synthesis off in the grafted cells. Morphometric analysis of the ovaries revealed that both groups grafted with GABA-producing cells had an increased incidence of large antral follicles (>500 micrometer) compared with animals grafted with native BAS-8.1 cells, but that within this category the incidence of steroidogenically more active follicles (i.e. larger than 600 micrometer) was greater in "GAD on" than in "GAD off" rats. These results indicate that a regionally discrete, temporally controlled increase in GABA availability to LHRH nerve terminals in the median eminence of the hypothalamus suffices to disrupt estrous cyclicity in the rat, and raise the possibility that similar local alterations in GABA homeostasis may contribute to the pathology of hypothalamic amenorrhea/oligomenorrhea in humans.


Asunto(s)
Estro , Hormona Liberadora de Gonadotropina/metabolismo , Eminencia Media/metabolismo , Tetraciclina/farmacología , Ácido gamma-Aminobutírico/biosíntesis , Animales , Línea Celular , Trasplante de Células , Estradiol/metabolismo , Femenino , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Hormona Luteinizante/metabolismo , Ratones , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley
19.
J Pediatr Endocrinol Metab ; 14(3): 245-56, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11308042

RESUMEN

Much effort has been devoted in recent years to unravel the neuroendocrine mechanisms responsible for the initiation of mammalian puberty. The concept that has emerged is that puberty results from the unfolding of a centrally originated process involving the concerted influence of neuronal systems that utilize excitatory and inhibitory amino acids as transmitters and astroglial networks that produce growth factors able to affect LHRH secretion. We discuss the idea that an isolated alteration of each of these components may result in the precocious activation of pulsatile LHRH release, and thus lead to idiopathic sexual precocity. According to this notion, such a premature activation of LHRH neuronal function would be neither associated with structural damage of the neuroendocrine brain system, nor related to a generalized activation of the neuronal-glial mechanisms underlying the onset of puberty. On the contrary, localized activation of discrete cellular subsets functionally connected to LHRH neurons would suffice to promote an increase in LHRH release of sufficient magnitude and duration to initiate the pubertal process.


Asunto(s)
Pubertad Precoz , Niño , Femenino , Hormona Liberadora de Gonadotropina/fisiología , Humanos
20.
J Clin Endocrinol Metab ; 86(1): 349-54, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11232023

RESUMEN

The presence of muscarinic receptors (MR) in the ovary of different species has been recognized, but the identity of these receptors as well as ovarian sources of their natural ligand, acetylcholine (ACh), have not been determined. Because luteinized human granulosa cells (GC) in culture express functional MR, we have determined whether the group of the related MR subtypes, M1R, M3R, and M5R, are present in vivo in human and rhesus monkey ovaries. To this end, ribonucleic acids (RNAs) of different human and monkey ovaries as well as RNAs from human GC and monkey oocytes were reverse transcribed and subjected to PCR amplification, followed by sequencing of the amplified complementary DNAs. Results obtained showed that M1R, M3R, and M5R messenger RNAs are present in adult human and monkey ovaries; oocytes express exclusively the M3R subtype, whereas GC express M1R and M5R. To determine the ovarian source(s) of the natural ligand of these ACh receptors, we attempted to localize the enzyme responsible for its synthesis with the help of a monoclonal antibody recognizing choline acetyltransferase for immunohistochemistry. In neither human nor monkey sections did we detect immunoreactive choline acetyltransferase-positive fibers or nerve cells, but, surprisingly, GC of antral follicles showed prominent staining. To determine whether GC can produce ACh, human cultured GC derived from preovulatory follicles were analyzed using a high pressure liquid chromatography technique. The results showed that these cells contained ACh in concentrations ranging from 4.2-11.5 pmol/10(6) cells. Samples of a rat granulosa cell line likewise contained ACh. Thus, the ovary contains multiple MR, and GC of antral follicles are able to synthesize ACh, the ligand of MR. We propose that ACh may serve as an as yet unrecognized factor involved in the complex regulation of ovarian function in the primate, e.g. regulation of cell proliferation or progesterone production.


Asunto(s)
Acetilcolina/biosíntesis , Ovario/metabolismo , Receptores Muscarínicos/metabolismo , Acetilcolina/metabolismo , Adulto , Animales , Encéfalo/metabolismo , Carnitina O-Acetiltransferasa/metabolismo , Células Cultivadas , Colina O-Acetiltransferasa/metabolismo , Femenino , Humanos , Inmunohistoquímica , Macaca mulatta , Persona de Mediana Edad , Datos de Secuencia Molecular , Filogenia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Mensajero/metabolismo , Receptores Muscarínicos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...