Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Front Neurosci ; 16: 744693, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35237119

RESUMEN

p140Cap, encoded by the gene SRCIN1 (SRC kinase signaling inhibitor 1), is an adaptor/scaffold protein highly expressed in the mouse brain, participating in several pre- and post-synaptic mechanisms. p140Cap knock-out (KO) female mice show severe hypofertility, delayed puberty onset, altered estrus cycle, reduced ovulation, and defective production of luteinizing hormone and estradiol during proestrus. We investigated the role of p140Cap in the development and maturation of the hypothalamic gonadotropic system. During embryonic development, migration of Gonadotropin-Releasing Hormone (GnRH) neurons from the nasal placode to the forebrain in p140Cap KO mice appeared normal, and young p140Cap KO animals showed a normal number of GnRH-immunoreactive (-ir) neurons. In contrast, adult p140Cap KO mice showed a significant loss of GnRH-ir neurons and a decreased density of GnRH-ir projections in the median eminence, accompanied by reduced levels of GnRH and LH mRNAs in the hypothalamus and pituitary gland, respectively. We examined the number of kisspeptin (KP) neurons in the rostral periventricular region of the third ventricle, the number of KP-ir fibers in the arcuate nucleus, and the number of KP-ir punctae on GnRH neurons but we found no significant changes. Consistently, the responsiveness to exogenous KP in vivo was unchanged, excluding a cell-autonomous defect on the GnRH neurons at the level of KP receptor or its signal transduction. Since glutamatergic signaling in the hypothalamus is critical for both puberty onset and modulation of GnRH secretion, we examined the density of glutamatergic synapses in p140Cap KO mice and observed a significant reduction in the density of VGLUT-ir punctae both in the preoptic area and on GnRH neurons. Our data suggest that the glutamatergic circuitry in the hypothalamus is altered in the absence of p140Cap and is required for female fertility.

2.
Artículo en Inglés | MEDLINE | ID: mdl-33013709

RESUMEN

Numerous chemicals derived from human activity are now disseminated in the environment where their exert estrogenic endocrine disrupting effects, and therefore represent major health concerns. The present study explored whether Methoxychlor (MXC), an insecticide with xenoestrogens activities, given during the perinatal period (from gestational day 11 to postnatal day 8) and at an environmentally dose [20 µg/kg (body weight)/day], would affect reproductive physiology and sexual behavior of the offspring in mice. While MXC exposure did not induce any differences in the weight gain of animals from birth to 4 months of age, a clear difference (although in opposite direction according to the sexes) was observed on the anogenital distance between intact and exposed animals. A similar effect was also observed on preputial separation and vaginal opening, which reflects, respectively, in males and females, puberty occurrence. The advanced puberty observed in females was associated with an enhanced expression of kisspeptin cells in the anteroventral periventricular region of the medial preoptic area. Exposure to MXC did not induce in adult females changes in the estrous cycle or in the weight of the female reproductive tract. By contrast, males showed reduced weight of the epididymis and seminiferous vesicles associated with reduced testosterone levels and seminiferous tubule diameter. We also showed that both males and females showed deficits in mate preference tests. As a whole, our results show that MXC impacts reproductive outcomes.


Asunto(s)
Disruptores Endocrinos/administración & dosificación , Insecticidas/administración & dosificación , Metoxicloro/administración & dosificación , Efectos Tardíos de la Exposición Prenatal/metabolismo , Reproducción/efectos de los fármacos , Conducta Sexual Animal/efectos de los fármacos , Animales , Ciclo Estral/efectos de los fármacos , Femenino , Kisspeptinas/metabolismo , Ratones , Embarazo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Área Preóptica/efectos de los fármacos , Área Preóptica/metabolismo , Maduración Sexual/efectos de los fármacos
3.
J Endocrinol ; 244(2): 273-283, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31671405

RESUMEN

Kiss1 neurons of the arcuate (ARC) nucleus form an interconnected network of cells that communicate via neurokinin B (encoded by Tac2) and its receptor (encoded by Tacr3) and play key roles in the control of the reproductive axis through sex hormone-regulated synthesis and release of kisspeptin peptides (Kp, encoded by Kiss1). The aim of this study was to determine whether the Kiss1 cell population of the ARC already displays sexually dimorphic features at embryonic age E16.5 in mice. At this time of development, Kiss1-GFP- and Kp-immunoreactive cell bodies were restricted to the ARC and not found in the pre-optic area (POA). The Kiss1-GFP cell population was identical in size between sexes but had significantly lower Kiss1, Tac2, and Tacr3 mRNA levels and lower Kp-ir fiber density in the POA in male compared to female fetuses. Receptors for androgen (Ar) and estrogen (Esr1, Esr2, Gpr30) and the Cyp19a1 gene (encoding the estradiol-producing enzyme aromatase) transcripts were also detected in fetal ARC Kiss1-GFP cells with significant sex differences for Ar (higher in males) and Esr1 (higher in females). Functional studies on primary cultures of sorted fetal Kiss1-GFP cells revealed a significant negative effect of estradiol treatment on neurite outgrowth on the fourth day of culture in the female group specifically. We conclude that the ARC Kiss1 cell population is already sexually differentiated at E16.5 and that its morphogenetic development may be particularly vulnerable to estradiol exposure at this early developmental time.


Asunto(s)
Estradiol/metabolismo , Kisspeptinas/metabolismo , Ratones/metabolismo , Neuritas/metabolismo , Caracteres Sexuales , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Kisspeptinas/genética , Masculino , Ratones/embriología , Ratones/genética , Ratones Endogámicos C57BL , Neuroquinina B/genética
4.
Chemosphere ; 221: 573-582, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30660913

RESUMEN

The present study evaluates the effects of adult exposure to low doses of a mixture of di-(2-ethylexyl)phthalate (DEHP) and nonylphenol (NP) on reproductive neuroendocrine function and behavior. The neural circuitry that processes male sexual behavior is tightly regulated by testosterone and its neural metabolite estradiol. In previous studies, we showed that adult exposure of mice to low doses of each of these widespread environmental contaminants resulted in altered sexual behavior, without any effect on the regulation of the gonadotropic axis. Here, adult C57BL/6J male mice were exposed to DEHP/NP (0.5 or 5 µg/kg body weight/day) for 4 weeks before starting the analyses. Mice treated with DEHP/NP at 0.5 µg/kg/day show altered olfactory preference, and fewer of them emit ultrasonic vocalization compared to the other treatment groups. These mice also exhibit a lower number of mounts and thrusts, increased locomotor activity and unaffected anxiety-state level, along with unaltered testosterone levels and kisspeptin system, a key regulator of the gonadotropic axis. Analysis of the neural circuitry that underlies sexual behavior showed that the number of cells expressing androgen and estrogen receptors is comparable between control and DEHP/NP-exposed males. The comparison of these data with those obtained in males exposed to each molecule separately highlights synergistic effects at the lower dose of contaminants of 0.5 µg/kg/day. In contrast, the effects previously observed for each molecule at 5 µg/kg/day were not detected. A detailed comparison of the effects triggered by separate or combined exposure to DEHP and NP is discussed.


Asunto(s)
Dietilhexil Ftalato/toxicidad , Contaminantes Ambientales/toxicidad , Sistemas Neurosecretores/efectos de los fármacos , Fenoles/toxicidad , Conducta Sexual Animal/efectos de los fármacos , Animales , Femenino , Kisspeptinas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Testosterona/metabolismo
5.
Sci Rep ; 8(1): 2988, 2018 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-29445187

RESUMEN

The neural circuitry processing male sexual behavior is tightly regulated by testosterone and its neural metabolite estradiol. The present study evaluated the effects of adult exposure to low doses of nonylphenol (NP), a widespread environmental contaminant, on the neuroendocrine regulation of testosterone and expression of sexual behavior. Oral exposure of C57BL/6J males to NP (0.5, 5 or 50 µg/kg/day) for 4 weeks did not affect circulating levels of testosterone or the kisspeptin system, a key regulator of the gonadotropic axis. In contrast, mice exposed to NP at 5 µg/kg/day emitted an increased number and duration of ultrasonic vocalizations, took longer to reach ejaculation and showed increased number of mounts, intromissions and thrusts. This was associated with normal olfactory preference and locomotor activity, and increased anxiety level. Analysis of the neural circuitry that underlies sexual behavior showed changes in the number of cells expressing androgen and estrogen receptors in males exposed to NP at 5 µg/kg/day. The neural circuitry underlying sexual behavior is thus highly sensitive to adult exposure to NP. Furthermore, almost all the observed effects were induced at 5 µg/kg/day of NP, indicating that this endocrine disrupter triggers a non-monotonic response in the adult male mouse brain.


Asunto(s)
Cortejo , Contaminantes Ambientales/metabolismo , Neuronas/fisiología , Fenoles/metabolismo , Vocalización Animal/fisiología , Animales , Ansiedad , Disruptores Endocrinos , Estradiol/metabolismo , Locomoción , Masculino , Ratones , Ratones Endogámicos C57BL , Conducción Nerviosa , Vías Olfatorias , Receptores Androgénicos/metabolismo , Receptores de Estrógenos/metabolismo , Testosterona/metabolismo
6.
Endocrinology ; 157(1): 304-22, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26562259

RESUMEN

Episodic release of GnRH is essential for reproductive function. In vitro studies have established that this episodic release is an endogenous property of GnRH neurons and that GnRH secretory pulses are associated with synchronization of GnRH neuron activity. The cellular mechanisms by which GnRH neurons synchronize remain largely unknown. There is no clear evidence of physical coupling of GnRH neurons through gap junctions to explain episodic synchronization. However, coupling of glial cells through gap junctions has been shown to regulate neuron activity in their microenvironment. The present study investigated whether glial cell communication through gap junctions plays a role in GnRH neuron activity and secretion in the mouse. Our findings show that Glial Fibrillary Acidic Protein-expressing glial cells located in the median eminence in close vicinity to GnRH fibers expressed Gja1 encoding connexin-43. To study the impact of glial-gap junction coupling on GnRH neuron activity, an in vitro model of primary cultures from mouse embryo nasal placodes was used. In this model, GnRH neurons possess a glial microenvironment and were able to release GnRH in an episodic manner. Our findings show that in vitro glial cells forming the microenvironment of GnRH neurons expressed connexin-43 and displayed functional gap junctions. Pharmacological blockade of the gap junctions with 50 µM 18-α-glycyrrhetinic acid decreased GnRH secretion by reducing pulse frequency and amplitude, suppressed neuronal synchronization and drastically reduced spontaneous electrical activity, all these effects were reversed upon 18-α-glycyrrhetinic acid washout.


Asunto(s)
Uniones Comunicantes/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/metabolismo , Mucosa Olfatoria/metabolismo , Células Receptoras Sensoriales/metabolismo , Animales , Biomarcadores/metabolismo , Células Cultivadas , Conexina 43/genética , Conexina 43/metabolismo , Embrión de Mamíferos/citología , Inhibidores Enzimáticos/farmacología , Uniones Comunicantes/efectos de los fármacos , Uniones Comunicantes/ultraestructura , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Hormona Liberadora de Gonadotropina/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Cinética , Eminencia Media/citología , Eminencia Media/efectos de los fármacos , Eminencia Media/metabolismo , Ratones Transgénicos , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Neuroglía/efectos de los fármacos , Neuroglía/ultraestructura , Neurotoxinas/farmacología , Mucosa Olfatoria/efectos de los fármacos , Mucosa Olfatoria/ultraestructura , Proteínas Recombinantes de Fusión/metabolismo , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/ultraestructura , Técnicas de Cultivo de Tejidos
7.
Hum Mol Genet ; 24(25): 7326-38, 2015 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-26464488

RESUMEN

Ovarian oestradiol is essential for pubertal maturation and adult physiology of the female reproductive axis. It acts at central and peripheral sites through two main oestrogen receptors (ER) α and ß. Here we investigate the role of ERß on central effects of oestradiol, by generating a mouse line specifically lacking the ERß gene in neuronal and glial cells. Central ERß deletion delays the age at vaginal opening and first oestrous and reduces uterine weight without affecting body growth. Analysis of factors necessary for pubertal progression shows reduced levels of Kiss1 transcripts at postnatal (P) day 25 in the preoptic area, but not in the mediobasal hypothalamus (MBH) of mutant females. In agreement with these data, the number of kisspeptin-immunoreactive neurons was decreased by 57-72% in the three subdivisions of the rostral periventricular area of the third ventricle (RP3V), whereas the density of kisspeptin-immunoreactive fibres was unchanged in the arcuate nucleus of mutant mice. These alterations do not involve changes in ERα mRNAs in the preoptic area and protein levels in the RP3V. The number and distribution of GnRH-immunoreactive cells were unaffected, but gonadotropin-releasing hormone (GnRH) transcript levels were higher in the P25 preoptic area of mutants. At adulthood, mutant females have normal oestrous cyclicity, kisspeptin system and exhibit unaltered sexual behaviour. They display, however, reduced ovary weight and increased anxiety-related behaviour during the follicular phase. This argues for the specific involvement of central ERß in the regulation of pubertal onset in female reproduction, possibly through prepubertal induction of kisspeptin expression in the RP3V.


Asunto(s)
Receptor beta de Estrógeno/metabolismo , Hormona Liberadora de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Animales , Estradiol/metabolismo , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/genética , Ciclo Estral/fisiología , Femenino , Hormona Liberadora de Gonadotropina/genética , Hipotálamo/metabolismo , Kisspeptinas/genética , Ratones , Pubertad/genética , Pubertad/metabolismo
8.
J Endocrinol ; 220(3): 375-88, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24403293

RESUMEN

Bisphenol A (BPA) is a widespread estrogenic compound. We investigated the effects of maternal exposure to BPA at reference doses on sexual behavior and neuroendocrine functions of female offspring in C57BL/6J mice. The dams were orally exposed to vehicle alone or vehicle-containing BPA at doses equivalent to the no observed adverse effect level (5 mg/kg body weight per day) and tolerable daily intake (TDI, 0.05 mg/kg body weight per day) level from gestational day 15 until weaning. Developmental exposure to BPA increased the lordosis quotient in naive females exposed to BPA at the TDI dose only. BPA exposure had no effect on olfactory preference, ability to express masculine behaviors or number of calbindin-positive cells, a sexually dimorphic population of the preoptic area. BPA at both doses selectively increased kisspeptin cell number in the preoptic periventricular nucleus of the rostral periventricular area of the third ventricle in adult females. It did not affect the number of GNRH-positive cells or percentage of kisspeptin appositions on GNRH neurons in the preoptic area. These changes were associated with higher levels of estradiol (E2) at the TDI dose while levels of LH, estrus cyclicity, ovarian and uterine weights, and fertility remained unaffected. Delay in the time of vaginal opening was observed during the postnatal period at TDI dose, without any alteration in body growth. This shows that developmental exposure to BPA at reference doses did not masculinize and defeminize the neural circuitry underlying sexual behavior in female mice. The TDI dose specifically exacerbated responses normally induced by ovarian E2, through estrogen receptor α, during the postnatal/prepubertal period.


Asunto(s)
Compuestos de Bencidrilo/efectos adversos , Exposición Materna/efectos adversos , Sistemas Neurosecretores/efectos de los fármacos , Fenoles/efectos adversos , Efectos Tardíos de la Exposición Prenatal/psicología , Animales , Estradiol/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Sistemas Neurosecretores/metabolismo , Ovario/crecimiento & desarrollo , Ovario/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/etiología , Efectos Tardíos de la Exposición Prenatal/metabolismo , Área Preóptica/efectos de los fármacos , Conducta Sexual Animal/efectos de los fármacos , Útero/crecimiento & desarrollo , Útero/metabolismo
9.
Endocrinology ; 155(2): 502-12, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24265451

RESUMEN

There are human reproduction concerns associated with extensive use of bisphenol A (BPA)-containing plastic and, in particular, the leaching of BPA into food and beverages. In this context, it remains unclear whether and how exposure to BPA interferes with the developmental organization and adult activation of male sexual behavior by testosterone. We evaluated the developmental and adult exposure to oral BPA at doses equivalent to the no-observed-adverse-effect-level (5 mg/kg body weight per day) and tolerable daily intake (TDI) (50 µg/kg body weight per day) on mouse sexual behavior and the potential mechanisms underlying BPA effects. Adult exposure to BPA reduced sexual motivation and performance at TDI dose only. Exposed males took longer to initiate mating and reach ejaculation despite normal olfactory chemoinvestigation. This deficiency was not restored by sexual experience and was associated with unchanged circulating levels of testosterone. By contrast, developmental exposure to BPA at TDI or no-observed-adverse-effect-level dose did not reduce sexual behavior or alter the neuroanatomical organization of the preoptic area. Disrupting the neural androgen receptor resulted in behavioral and neuroanatomical effects similar to those induced by adult exposure to TDI dose. Moreover, adult exposure of mutant males to BPA at TDI dose did not trigger additional alteration of sexual behavior, suggesting that BPA and neural androgen receptor mutation share a common mechanism of action. This shows, for the first time, that the neural circuitry underlying male sexual behavior is vulnerable to chronic adult exposure to low dose of BPA and suggests that BPA could act in vivo as an antiandrogenic compound.


Asunto(s)
Compuestos de Bencidrilo/administración & dosificación , Estrógenos no Esteroides/administración & dosificación , Red Nerviosa/efectos de los fármacos , Fenoles/administración & dosificación , Área Preóptica/efectos de los fármacos , Conducta Sexual Animal/efectos de los fármacos , Administración Oral , Animales , Conducta de Elección/efectos de los fármacos , Receptor alfa de Estrógeno/metabolismo , Conducta Exploratoria/efectos de los fármacos , Kisspeptinas/metabolismo , Masculino , Ratones , Red Nerviosa/metabolismo , Área Preóptica/metabolismo , Receptores Androgénicos/metabolismo , Olfato/efectos de los fármacos
10.
Peptides ; 45: 85-90, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23651990

RESUMEN

Kiss1 mRNA and its corresponding peptide products, kisspeptins, are expressed in two restricted brain areas of rodents, the anteroventral periventricular nucleus (AVPV) and the arcuate nucleus (ARC). The concentration of mature kisspeptins may not directly correlate with Kiss1 mRNA levels, because mRNA translation and/or posttranslational modification, degradation, transportation and release of kisspeptins could be regulated independently of gene expression, and there may thus be differences in kisspeptin expression even in species with similar Kiss1 mRNA profiles. We measured and compared kisspeptin-immunoreactivity in both nuclei and both sexes of rats and mice and quantified kisspeptin-immunoreactive nerve fibers. We also determined Kiss1 mRNA levels and measured kisspeptin-immunoreactivity in colchicine pretreated rats. Overall, we find higher levels of kisspeptin-immunoreactivity in the mouse compared to the rat, independently of brain region and gender. In the female mouse AVPV high numbers of kisspeptin-immunoreactive neurons were present, while in the rat, the female AVPV displays a similar number of kisspeptin-immunoreactive neurons compared to the level of Kiss1 mRNA expressing cells, only after axonal transport inhibition. Interestingly, the density of kisspeptin innervation in the anterior periventricular area was higher in female compared to male in both species. Species differences in the ARC were evident, with the mouse ARC containing dense fibers, while the rat ARC contains clearly discernable cells. In addition, we show a marked sex difference in the ARC, with higher kisspeptin levels in females. These findings show that the translation of Kiss1 mRNA and/or the degradation/transportation/release of kisspeptins are different in mice and rats.


Asunto(s)
Kisspeptinas/genética , ARN Mensajero/genética , Animales , Núcleos Talámicos Anteriores/citología , Núcleos Talámicos Anteriores/efectos de los fármacos , Núcleos Talámicos Anteriores/metabolismo , Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/metabolismo , Colchicina/farmacología , Femenino , Regulación de la Expresión Génica , Inmunohistoquímica , Kisspeptinas/metabolismo , Masculino , Ratones , Fibras Nerviosas/efectos de los fármacos , Fibras Nerviosas/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Biosíntesis de Proteínas , Transporte de Proteínas , ARN Mensajero/metabolismo , Ratas , Factores Sexuales , Especificidad de la Especie
11.
Endocrinology ; 152(1): 214-22, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21047947

RESUMEN

We tested the working hypothesis that Fos will identify the critical population of kisspeptin neurons that accompanies the LHRH surge using a synchronized follicular phase model in intact cycling ewes. The model generates an LH surge that starts within a defined 2-h window in a 20-d synchronized cycle. With a modified push-pull cannula in vivo LHRH release from the median eminence was sampled in luteal phase ewes, ewes undergoing an LH surge for 2-4 h, and postsurge animals whose LH surge peaked 10-12 h earlier. In vivo release of LHRH was lower in the luteal and follicular phases than in animals undergoing an LH surge (P < 0.01); it fell to presurge levels after the LH surge. Ewes killed 2-4 h after the surge started, expressed Fos in a large portion of preoptic area (POA) kisspeptin (53.90 ± 4.69%, P < 0.01) and LHRH neurons (48.20 ± 4.49%, P < 0.0001) compared with animals euthanized at any of the other times tested (under <5% of the cells activated). Little Fos activation (under 5%) was observed during any of the times sampled in arcuate (Arc) kisspeptin neurons. The relationship between the number of LHRH neurons and the POA kisspeptin neurons stimulated showed a striking positive correlation with r(2) = 0.68, P = 0.0003, reinforcing the evidence that POA kisspeptin neurons actively participate in the stimulation of LHRH surges.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Hormona Luteinizante/metabolismo , Eminencia Media/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ovinos/fisiología , Proteínas Supresoras de Tumor/metabolismo , Animales , Regulación de la Expresión Génica/fisiología , Neuronas/fisiología , Área Preóptica , Proteínas Supresoras de Tumor/genética
12.
J Biol Rhythms ; 26(6): 486-96, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22215607

RESUMEN

To adapt to seasonal variations in the environment, most mammalian species exhibit seasonal cycles in their physiology and behavior. Seasonal plasticity in the structure and function of the central nervous system contributes to the adaptation of this physiology in seasonal mammals. As part of these plasticity mechanisms, seasonal variations in proliferation rate and neuron production have been extensively studied in songbirds. In this report, we investigated whether this type of brain plasticity also occurs in sheep, a seasonal species, by assessing variations in cell proliferation in the sheep diencephalon. We administered the cell birth marker 5'-bromodeoxyuridine (BrdU) to adult female sheep in July and December, during long and short photoperiod, respectively. The BrdU incorporation was analyzed and quantified in the hypothalamus, a key center for neuroendocrine regulations, as well as in other structures involved in relaying neuroendocrine and sensory information, including the median eminence, the pars tuberalis of the pituitary gland, and the thalamus. In December, 2-fold and 6-fold increases in the number of BrdU+ nuclei were observed in the hypothalamus and thalamus, respectively, when compared with July. This variation is independent of the influence of peripheral gonadal estradiol variations. An inverse seasonal regulation of cell proliferation was observed in the pars tuberalis. In contrast, no seasonal variation in cell proliferation was seen in the subventricular zone of the lateral ventricle. Many of the newborn cells in the adult ovine hypothalamus and thalamus differentiate into neurons and glial cells, as assessed by the expression of neuronal (DCX, NeuN) and glial (GFAP, S100B) fate markers. In summary, we show that the estimated cell proliferation rates in the sheep hypothalamus, thalamus, and pars tuberalis are different between seasons. These variations are independent of the seasonal fluctuations of peripheral estradiol levels, unlike the results described in the brain nuclei involved in song control of avian species.


Asunto(s)
Ciclos de Actividad/fisiología , Encéfalo/anatomía & histología , Encéfalo/fisiología , Proliferación Celular , Adenohipófisis/anatomía & histología , Adenohipófisis/fisiología , Estaciones del Año , Animales , Biomarcadores/metabolismo , Bromodesoxiuridina/metabolismo , Femenino , Hipotálamo/citología , Hipotálamo/metabolismo , Plasticidad Neuronal/fisiología , Neuronas/citología , Neuronas/fisiología , Fotoperiodo , Ovinos
13.
Eur J Neurosci ; 32(12): 2031-41, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21143658

RESUMEN

The neurohormone gonadotropin-releasing hormone (GnRH) is critical for all the aspects of reproductive life in vertebrates. GnRH is secreted by a small number of neurons dispersed within the preoptic-hypothalamic region. These neurons are derived from the embryonic olfactory pit. They then migrate along olfactory, vomeronasal and terminal nerves to their final destination. Classical approaches to study the regulation of GnRH secretion during the reproductive cycle have focused on the various neuronal inputs on GnRH neurons and their regulation by ovarian steroids. However, it is well known that steroids will change the microenvironment of neuronal networks and can induce plasticity and functional changes. In this review, we will focus on the intimate relationship of developing and adult GnRH neurons with the polysialylated form of neural cell adhesion molecule (PSA-NCAM), a major molecular actor in the morphogenesis and adult plasticity of the nervous system. We will first recapitulate the spatiotemporal relationship between PSA-NCAM and migrating GnRH neurons during embryogenesis of various vertebrate species and discuss its importance for GnRH neuron development as shown by various loss of function studies. In the adult, we will review the relationships between PSA-NCAM and GnRH neurons across various physiological states, and open the discussion to the use of new model systems that can help to unravel the function and mechanism of action of PSA-NCAM on GnRH neuronal network activity and GnRH release.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Plasticidad Neuronal/fisiología , Neuronas/fisiología , Ácidos Siálicos/metabolismo , Animales , Movimiento Celular , Humanos , Hipotálamo/anatomía & histología , Hipotálamo/crecimiento & desarrollo , Hipotálamo/fisiología , Modelos Neurológicos , Neuronas/citología
14.
Eur J Neurosci ; 32(12): 2042-52, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21143659

RESUMEN

In adult mammalian brain, two main germinative regions located in the subventricular zone of the lateral ventricle and in the subgranular cell layer of the hippocampal dentate gyrus have been considerably documented and are still under intense scrutiny. However, new neuron formation has recently been reported in various other brain areas including the hypothalamus. This central structure, responsible for the control of many major neuroendocrine functions such as reproduction, expresses high levels of PSA-NCAM and nestin, both proteins being involved in structural and morphological plasticity mechanisms. Cell proliferation and new neuron production have been demonstrated in the adult hypothalamus of numerous species, although not hitherto described in non-human primates and humans. Similarly to the subventricular zone and in the subgranular cell layer, the adult hypothalamic neurogenesis process is subject to dynamic regulation by various physiological and pharmacological signals. Several pieces of evidence support the hypothesis that a stem cell niche-like architecture exist in the hypothalamus region lining the third ventricle thereby enabling adult neural stem cells to continuously generate neurons in vivo throughout life. Furthermore, recent data indicating that new hypothalamic neurons may become functionally implicated in sensory information processing endorse the assumption that the hypothalamus might be a neurogenic region.


Asunto(s)
Encéfalo/anatomía & histología , Encéfalo/fisiología , Hipotálamo/citología , Hipotálamo/fisiología , Neurogénesis/fisiología , Tercer Ventrículo/citología , Tercer Ventrículo/fisiología , Animales , Biomarcadores/metabolismo , Proliferación Celular , Humanos , Plasticidad Neuronal/fisiología , Nicho de Células Madre
15.
J Soc Biol ; 203(1): 19-28, 2009.
Artículo en Francés | MEDLINE | ID: mdl-19358808

RESUMEN

Reproduction in mammals is directly controlled by GnRH neurons. These neurons are regulated by many external and internal factors, among which sexual steroids, in particular oestradiol, play an important part. However the mechanisms through which these steroids regulate GnRH secretion are largely unappreciated, and the neurochemical identity of central neurons liable to transmit the steroidal information to GnRH neurons is not completely clarified. Many functional neuroanatomy studies have been carried out on the ovine model, which is particularly favorable to understand the neuroendocrine mechanisms controlling reproduction. These studies have brought about the identification of some of the potential actors in this regulation. The present review reports the major results concerning two recently discovered neuropeptides, galanin and kisspeptin, which appear to be major actors in integration of signals regulating reproduction, among which steroids. These results have revealed the major interaction sites between neurons expressing these neuropeptides and GnRH neurons.


Asunto(s)
Galanina/fisiología , Hormona Liberadora de Gonadotropina/fisiología , Hipotálamo/fisiología , Neuronas/fisiología , Neuropéptidos/fisiología , Ovinos/anatomía & histología , Animales , Estradiol/fisiología , Femenino , Hipotálamo/anatomía & histología , Kisspeptinas , Ratones , Ratones Noqueados , Primates/fisiología , Proteínas/genética , Proteínas/fisiología , Reproducción/fisiología , Roedores/fisiología , Especificidad de la Especie
16.
Stem Cells ; 25(12): 3016-25, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17823239

RESUMEN

The development of stem cell-based neural repair strategies requires detailed knowledge on the interaction of migrating donor cells with the host brain environment. Here we report that overexpression of polysialic acid (PSA), a carbohydrate polymer attached to the neural cell adhesion molecule (NCAM), in embryonic stem (ES) cell-derived glial precursors (ESGPs) strikingly modifies their migration behavior in response to guidance cues. ESGPs transduced with a retrovirus encoding the polysialyltransferase STX exhibit enhanced migration in monolayer cultures and an increased penetration of organotypic slice cultures. Chemotaxis assays show that overexpression of PSA results in an enhanced chemotactic migration toward gradients of a variety of chemoattractants, including fibroblast growth factor 2 (FGF2), platelet-derived growth factor, and brain-derived neurotrophic factor (BDNF), and that this effect is mediated via the phosphatidylinositol 3'-kinase (PI3K) pathway. Moreover, PSA-overexpressing ESGPs also exhibit an enhanced chemotactic response to tissue explants derived from different brain regions. The effect of polysialylation on directional migration is preserved in vivo. Upon transplantation into the adult striatum, PSA-overexpressing but not control cells display a targeted migration toward the subventricular zone. On the basis of these data, we propose that PSA plays a crucial role in modulating the ability of migrating precursor cells to respond to regional guidance cues within the brain tissue. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Quimiotaxis/fisiología , Señales (Psicología) , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Neuronas/citología , Neuronas/metabolismo , Ácidos Siálicos/metabolismo , Animales , Células Cultivadas , Células Madre Embrionarias/química , Ratones , Moléculas de Adhesión de Célula Nerviosa/fisiología , Neuronas/química , Ratas , Ácidos Siálicos/fisiología
17.
Proc Natl Acad Sci U S A ; 104(25): 10714-9, 2007 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-17563351

RESUMEN

The G protein-coupled receptor GPR54 (AXOR12, OT7T175) is central to acquisition of reproductive competency in mammals. Peptide ligands (kisspeptins) for this receptor are encoded by the Kiss1 gene, and administration of exogenous kisspeptins stimulates hypothalamic gonadotropin-releasing hormone (GnRH) release in several species, including humans. To establish that kisspeptins are the authentic agonists of GPR54 in vivo and to determine whether these ligands have additional physiological functions we have generated mice with a targeted disruption of the Kiss1 gene. Kiss1-null mice are viable and healthy with no apparent abnormalities but fail to undergo sexual maturation. Mutant female mice do not progress through the estrous cycle, have thread-like uteri and small ovaries, and do not produce mature Graffian follicles. Mutant males have small testes, and spermatogenesis arrests mainly at the early haploid spermatid stage. Both sexes have low circulating gonadotropin (luteinizing hormone and follicle-stimulating hormone) and sex steroid (beta-estradiol or testosterone) hormone levels. Migration of GnRH neurons into the hypothalamus appears normal with appropriate axonal connections to the median eminence and total GnRH content. The hypothalamic-pituitary axis is functional in these mice as shown by robust luteinizing hormone secretion after peripheral administration of kisspeptin. The virtually identical phenotype of Gpr54- and Kiss1-null mice provides direct proof that kisspeptins are the true physiological ligand for the GPR54 receptor in vivo. Kiss1 also does not seem to play a vital role in any other physiological processes other than activation of the hypothalamic-pituitary-gonadal axis, and loss of Kiss1 cannot be overcome by compensatory mechanisms.


Asunto(s)
Hormona Liberadora de Gonadotropina/metabolismo , Hipogonadismo/genética , Hipogonadismo/metabolismo , Proteínas/genética , Envejecimiento , Animales , Femenino , Marcación de Gen , Hormona Liberadora de Gonadotropina/análisis , Kisspeptinas , Masculino , Ratones , Ratones Mutantes
18.
Glia ; 53(8): 868-78, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16598779

RESUMEN

Schwann cells, the myelin-forming cells of the PNS, are attractive candidates for remyelination therapy as they can remyelinate CNS axons. Yet their integration in CNS tissue appears hampered, at least in part, by their limited motility in the CNS environment. As the polysialylated (PSA) form of NCAM regulates migration of neural precursors in the CNS and is not expressed by developing Schwann cells, we investigated whether conferring sustained expression of PSA to Schwann cells derived from postnatal rats enhances their motility. Cells were transduced with a retrovirus encoding polysialyl-transferase STX, an enzyme that synthesizes PSA on NCAM. Migration of wild type and transduced cells expressing STX or the marker gene alkaline phosphatase was examined using a gap bridging assay in dissociated cells and by grafting cells in slice cultures of postnatal brain. Migration of PSA expressing cells was significantly increased in both models, as compared to control cells, and this effect was abolished by endoneuraminidase-N stripping of PSA. PSA-positive Schwann cells retained the ability to differentiate in vitro and expressed the Krox20 and P zero myelination markers. When grafted in neonatal cerebellar slices, STX-transduced cells started to myelinate Purkinje cell axons like control cells and make myelin internodes after 2 to 3 weeks. PSA was redistributed on the cell membrane and downregulated during differentiation in pure Schwann cell cultures and slice co-cultures. Thus, migratory properties of PNS myelin-forming cells within the CNS can be enhanced without altering their differentiation program. This finding may be beneficial for the development of remyelination therapies.


Asunto(s)
Movimiento Celular/fisiología , Vaina de Mielina/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Nervios Periféricos/metabolismo , Células de Schwann/metabolismo , Ácidos Siálicos/biosíntesis , Ácidos Siálicos/metabolismo , Animales , Animales Recién Nacidos , Axones/fisiología , Axones/ultraestructura , Biomarcadores/metabolismo , Comunicación Celular/fisiología , Diferenciación Celular/genética , Células Cultivadas , Regulación hacia Abajo/fisiología , Proteína 2 de la Respuesta de Crecimiento Precoz/metabolismo , Ingeniería Genética/métodos , Vectores Genéticos , Proteína P0 de la Mielina/metabolismo , Vaina de Mielina/ultraestructura , Regeneración Nerviosa/fisiología , Técnicas de Cultivo de Órganos , Nervios Periféricos/citología , Ratas , Células de Schwann/citología , Células de Schwann/trasplante , Ácidos Siálicos/genética , Sialiltransferasas/genética , Transducción Genética/métodos , Regulación hacia Arriba/fisiología
19.
Mol Cell Neurosci ; 27(2): 151-62, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15485771

RESUMEN

Polysialic acid (PSA) on NCAM is an important modulator of cell-cell interactions during development and regeneration. Here we investigated whether PSA overexpression influences neural cell migration and myelination. We stably expressed a GFP-tagged polysialytransferase, PSTGFP, in mouse neurospheres and induced prolonged PSA synthesis. Using a chick xenograft assay for migration, we show that PSA can instruct precursor migration along the ventral pathway. PSA persistence did not change neural precursor multipotentiality in vitro but induced a delay in oligodendrocyte differentiation. PSTGFP+ precursors showed widespread engraftment in shiverer brain, closely similar to that observed with control precursors expressing a fluorescent protein. Initially, myelination by oligodendrocytes was delayed but, eventually, down-regulation of PSTGFP occurred, allowing myelination to proceed. Thus down-regulation of polysialyltransferases takes place even in cells where its RNA is under the control of a heterologous promoter and engineering PSA overexpression in neural precursors does not cause irreversible unphysiological effects.


Asunto(s)
Movimiento Celular/fisiología , Fibras Nerviosas Mielínicas/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/biosíntesis , Neuronas/metabolismo , Ácidos Siálicos/biosíntesis , Células Madre/metabolismo , Células 3T3 , Animales , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Embrión de Pollo , Regulación de la Expresión Génica/fisiología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fibras Nerviosas Mielínicas/trasplante , Molécula L1 de Adhesión de Célula Nerviosa/genética , Neuronas/trasplante , Ingeniería de Proteínas/métodos , Ácidos Siálicos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA