Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros











Intervalo de año de publicación
1.
Domest Anim Endocrinol ; 62: 1-9, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28843181

RESUMEN

Prenatal androgens are largely responsible for growth and differentiation of the genital tract and testis and for organization of the control mechanisms regulating male reproductive physiology and behavior. The aim of the present study was to evaluate the impact of inappropriate exposure to excess testosterone (T) during the first trimester of fetal development on the reproductive function, sexual behavior, and fertility potential of rams. We found that biweekly maternal T propionate (100 mg) treatment administered from Day 30-58 of gestation significantly decreased (P < 0.05) postpubertal scrotal circumference and sperm concentration. Prenatal T exposure did not alter ejaculate volume, sperm motility and morphology or testis morphology. There was, however, a trend for more T-exposed rams than controls to be classified as unsatisfactory potential breeders during breeding soundness examinations. Postnatal serum T concentrations were not affected by prenatal T exposure, nor was the expression of key testicular genes essential for spermatogenesis and steroidogenesis. Basal serum LH did not differ between treatment groups, nor did pituitary responsiveness to GnRH. T-exposed rams, like control males, exhibited vigorous libido and were sexually attracted to estrous females. In summary, these results suggest that exposure to exogenous T during the first trimester of gestation can negatively impact spermatogenesis and compromise the reproductive fitness of rams.


Asunto(s)
Efectos Tardíos de la Exposición Prenatal , Conducta Sexual Animal/efectos de los fármacos , Ovinos/fisiología , Recuento de Espermatozoides/veterinaria , Testículo/efectos de los fármacos , Testosterona/farmacología , Animales , Femenino , Hormona Liberadora de Gonadotropina/farmacología , Hormona Luteinizante/metabolismo , Masculino , Embarazo , Caracteres Sexuales , Espermatogénesis/efectos de los fármacos , Testículo/crecimiento & desarrollo , Testosterona/administración & dosificación
2.
J Neuroendocrinol ; 28(7)2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27262161

RESUMEN

This review highlights the principal effects of steroid hormones at central and peripheral levels in the neuroendocrine axis. The data discussed highlight the principal role of oestrogens and testosterone in hormonal programming in relation to sexual orientation, reproductive and metabolic programming, and the neuroendocrine mechanism involved in the development of polycystic ovary syndrome phenotype. Moreover, consistent with the wide range of processes in which steroid hormones take part, we discuss the protective effects of progesterone on neurodegenerative disease and the signalling mechanism involved in the genesis of oestrogen-induced pituitary prolactinomas.


Asunto(s)
Estrógenos/fisiología , Metabolismo/fisiología , Progesterona/fisiología , Reproducción/fisiología , Conducta Sexual/fisiología , Médula Espinal/fisiopatología , Testosterona/fisiología , Animales , Carcinogénesis , Humanos , Enfermedades Neurodegenerativas/fisiopatología , Factores Protectores
3.
J Neuroendocrinol ; 28(5)2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27005749

RESUMEN

Testosterone exposure during midgestation differentiates neural circuits controlling sex-specific behaviours and patterns of gonadotrophin secretion in male sheep. Testosterone acts through androgen receptors (AR) and/or after aromatisation to oestradiol and binding to oestrogen receptors. The present study assessed the role of AR activation in male sexual differentiation. We compared rams that were exposed to the AR antagonist flutamide (Flu) throughout the critical period (i.e. days 30-90 of gestation) to control rams and ewes that received no prenatal treatments. The external genitalia of all Flu rams were phenotypically female. Testes were positioned s.c. in the inguinal region of the abdomen, exhibited seasonally impaired androgen secretion and were azospermic. Flu rams displayed male-typical precopulatory and mounting behaviours but could not intromit or ejaculate because they lacked a penis. Flu rams exhibited greater mounting behaviour than control rams and, similar to controls, showed sexual partner preferences for oestrous ewes. Neither control, nor Flu rams responded to oestradiol treatments with displays of female-typical receptive behaviour or LH surge responses, whereas all control ewes responded as expected. The ovine sexually dimorphic nucleus in Flu rams was intermediate in volume between control rams and ewes and significantly different from both. These results indicate that prenatal anti-androgen exposure is not able to block male sexual differentiation in sheep and suggest that compensatory mechanisms intervene to maintain sufficient androgen stimulation during development.


Asunto(s)
Antagonistas de Receptores Androgénicos/administración & dosificación , Flutamida/administración & dosificación , Hormonas Esteroides Gonadales/metabolismo , Receptores Androgénicos/fisiología , Diferenciación Sexual , Conducta Sexual Animal , Animales , Estradiol/administración & dosificación , Femenino , Hormona Liberadora de Gonadotropina/administración & dosificación , Hormona Luteinizante/metabolismo , Masculino , Embarazo , Diferenciación Sexual/efectos de los fármacos , Conducta Sexual Animal/efectos de los fármacos , Oveja Doméstica , Testículo/citología , Testículo/efectos de los fármacos , Testosterona/metabolismo
4.
J Neuroendocrinol ; 26(5): 321-8, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24730418

RESUMEN

Gonadal steroid hormones play important roles during critical periods of development to organise brain structures that control sexually dimorphic neuroendocrine responses and behaviours. Specific receptors for androgens and oestrogens must be expressed at appropriate times during development to mediate these processes. The present study was performed to test for sex differences in the relative expression of oestrogen receptor (ER)α and androgen receptor (AR) mRNA during the window of time in gestation that is critical for behavioural masculinisation and differentiation of the ovine sexually dimorphic nucleus (oSDN) in the sheep. In addition, we examined whether ERα and AR mRNA expression is localised within the nascent oSDN and could be involved in its development. Using the quantitative real-time polymerase chain reaction, we found that females expressed more ERα mRNA than males in medial preoptic area and medial basal hypothalamus during the mid-gestational critical period for brain sexual differentiation. No sex differences were found for AR mRNA in any tissue examined or for ERα in amygdala and frontal cortex. Using radioactive in situ hybridisation, we found that the distributions of ERα and AR mRNA overlapped with aromatase mRNA, which delineates the boundaries of the developing oSDN and identifies this nucleus as a target for both androgens and oestrogens. These data demonstrate that the transcriptional machinery for synthesising gonadal steroid receptors is functional in the foetal lamb brain during the critical period for sexual differentiation and suggest that possible mechanisms for establishing dimorphisms controlled by gonadal steroids may exist at the level of steroid hormone receptor expression.


Asunto(s)
Química Encefálica/fisiología , Receptor alfa de Estrógeno/biosíntesis , Feto/metabolismo , ARN Mensajero/biosíntesis , Receptores Androgénicos/biosíntesis , Adulto , Animales , Femenino , Desarrollo Fetal/fisiología , Humanos , Masculino , Datos de Secuencia Molecular , Embarazo , Caracteres Sexuales , Diferenciación Sexual/fisiología , Ovinos
5.
J Neuroendocrinol ; 22(6): 518-26, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20236232

RESUMEN

Developmental exposure to the agricultural fungicide vinclozolin can impair reproductive function in male rabbits and was previously found to decrease the number of immunoreactive-gonadotrophin-releasing hormone (GnRH) neurones in the region of the organum vasculosum of the lamina terminalis and rostral preoptic area by postnatal week (PNW) 6. In the present study, in an aim to further examine the disruption of GnRH neurones by foetal vinclozolin exposure, pregnant rabbits were dosed orally with vinclozolin, flutamide or carrot paste vehicle for the last 2 weeks of gestation. Offspring were euthanised at birth (males and females), PNW 6 (females), PNW 26 (adult males) or PNW 30 (adult females) of age. At birth and in adults, brains were sectioned and processed for immunoreactive GnRH. The numbers of immunoreactive GnRH neuronal perikarya were significantly decreased in vinclozolin-treated rabbits at birth and in adult littermates. By contrast, there was an increase in GnRH immunoreactivity in the terminals in the region of the median eminence. Analysis of PNW 6 female brains by radioimmunoassay revealed a two-fold increase in GnRH peptide content in the mediobasal hypothalamus in vinclozolin-treated rabbits. This finding was complemented by immunofluorescence analyses, which revealed a 2.8-fold increase in GnRH immunoreactivity in the median eminence of vinclozolin compared to vehicle-treated females at PNW 30. However, there was no difference between treatment groups in the measures of reproduction that were evaluated: ejaculation latency, conception rates or litter size. These results indicate that sub-acute, prenatal vinclozolin treatment is sufficient to create perdurable alterations in the GnRH neuronal network that forms an important input into the reproductive axis. Finally, the effect of vinclozolin on the GnRH neuronal network was not comparable to that of flutamide, suggesting that vinclozolin was not acting through anti-androgenic mechanisms.


Asunto(s)
Fungicidas Industriales/farmacología , Hormona Liberadora de Gonadotropina/metabolismo , Neuronas/efectos de los fármacos , Oxazoles/farmacología , Efectos Tardíos de la Exposición Prenatal , Animales , Femenino , Inmunohistoquímica , Masculino , Microscopía Fluorescente , Neuronas/metabolismo , Embarazo , Conejos , Radioinmunoensayo , Reproducción
6.
Biol Reprod ; 81(2): 388-95, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19403926

RESUMEN

Domestic pigs have three CYP19 genes encoding functional paralogues of the enzyme aromatase cytochrome P450 (P450arom) that are expressed in the gonads, placenta, and preimplantation blastocyst. All catalyze estrogen synthesis, but the gonadal-type enzyme is unique in also synthesizing a nonaromatizable biopotent testosterone metabolite, 1OH-testosterone (1OH-T). P450arom is expressed in the vertebrate brain, is higher in males than females, but has not been investigated in pigs, to our knowledge. Therefore, these studies defined which of the porcine CYP19 genes was expressed, and at what level, in adult male and female hypothalamus. Regional expression was examined in mature boars, and regulation of P450arom expression in neonatal boars was investigated by inhibition of P450arom with letrozole, which is known to reprogram testicular expression. Pig hypothalami expressed the gonadal form of P450arom (redesignated the "gonadal/hypothalamic" porcine CYP19 gene and paralogue) based on functional analysis confirmed by cloning and sequencing transcripts. Hypothalamic tissue synthesized 1OH-T and was sensitive to the selective P450arom inhibitor etomidate. Levels were 4-fold higher in male than female hypothalami, with expression in the medial preoptic area and lateral borders of the ventromedial hypothalamus of boars. In vivo, letrozole-treated neonates had increased aromatase activity in hypothalami but decreased activity in testes. Therefore, although the same CYP19 gene is expressed in both tissues, expression is regulated differently in the hypothalamus than testis. These investigations, the first such studies in pig brain to our knowledge, demonstrate unusual aspects of P450arom expression and regulation in the hypothalamus, offering promise of gaining better insight into roles of P450arom in reproductive function.


Asunto(s)
Inhibidores de la Aromatasa/farmacología , Aromatasa/metabolismo , Etomidato/farmacología , Hipotálamo/enzimología , Nitrilos/farmacología , Sus scrofa/metabolismo , Triazoles/farmacología , Análisis de Varianza , Animales , Aromatasa/química , Aromatasa/genética , Inhibidores de la Aromatasa/metabolismo , Secuencia de Bases , Estradiol/sangre , Femenino , Regulación Enzimológica de la Expresión Génica , Gónadas/enzimología , Hipotálamo/anatomía & histología , Hipotálamo/efectos de los fármacos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Letrozol , Masculino , Microsomas/efectos de los fármacos , Microsomas/enzimología , Microsomas/metabolismo , Datos de Secuencia Molecular , Hipófisis/enzimología , Placenta/enzimología , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Caracteres Sexuales , Estadísticas no Paramétricas , Sus scrofa/crecimiento & desarrollo , Testículo/efectos de los fármacos , Testículo/enzimología , Testosterona/sangre
7.
J Neuroendocrinol ; 21(4): 359-64, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19207819

RESUMEN

In our laboratory, the domestic ram is used as an experimental model to study the early programming of neural mechanisms underlying same-sex partner preference. This interest developed from the observation that approximately 8% of domestic rams are sexually attracted to other rams (male-oriented) in contrast to the majority of rams that are attracted to oestrous ewes (female-oriented). One prominent feature of sexual differentiation in many species is the presence of a sexually dimorphic nucleus (SDN) in the preoptic/anterior hypothalamus that is larger in males than in females. Lesion studies in rats and ferrets implicate the SDN in the expression of sexual preferences. We discovered an ovine SDN (oSDN) in the preoptic/anterior hypothalamus that is smaller in male- than in female-oriented rams and similar in size to the oSDN of ewes. Neurones of the oSDN show abundant aromatase expression that is also reduced in male-oriented compared to female-oriented rams. This observation suggests that sexual partner preferences are neurologically hard-wired and could be influenced by hormones. Aromatase-containing neurones constitute a nascent oSDN as early as day 60 of gestation, which becomes sexually dimorphic by day 135 of gestation when it is two-fold larger in males than in females. Exposure of fetal female lambs to exogenous testosterone from days 30-90 of gestation resulted in a masculinised oSDN. These data demonstrate that the oSDN develops prenatally and may influence adult sexual preferences. Surprisingly, inhibition of aromatase activity in the brain of ram foetuses during the critical period did not interfere with defeminisation of adult sexual partner preference or oSDN volume. These results fail to support an essential role for neural aromatase in the sexual differentiation of sheep brain and behaviour. Thus, we propose that oSDN morphology and male-typical partner preferences may instead be programmed through an androgen receptor mechanism not involving aromatisation.


Asunto(s)
Hipotálamo/embriología , Hipotálamo/fisiología , Preferencia en el Apareamiento Animal/fisiología , Neuronas/fisiología , Ovinos/embriología , Animales , Aromatasa/metabolismo , Estrógenos/metabolismo , Femenino , Homosexualidad Masculina , Hipotálamo/anatomía & histología , Masculino , Tamaño de los Órganos , Caracteres Sexuales , Ovinos/fisiología , Esteroides/sangre , Testosterona/metabolismo
8.
Reproduction ; 135(5): 733-8, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18304985

RESUMEN

Two experiments were conducted to determine whether an estradiol challenge could cause a female-type LH surge in castrated male- and female-oriented rams (MORs and FORs). Administration of 17beta-estradiol to castrated MORs and FORs and ovariectomized ewes caused an initial reduction in LH secretion followed for 12-20 h by a surge release of LH in the ewes. No surge release of LH occurred in the MORs and FORs. The pattern of changes in LH secretion within rams and ewes did not differ between the breeding and nonbreeding seasons. Treatment failed to elicit female-typical receptive sexual behaviors in the rams but did stimulate increased sexual receptivity in the ewes as determined by the measures of responsiveness to the teaser ram. Overall, no differences were found in hypothalamic-hypophyseal function in response to exogenous estradiol between MORs and FORs. These data are interpreted to suggest that in contrast to sexual attraction, the neural mechanisms controlling the LH surge and female receptivity are defeminized in MORs.


Asunto(s)
Estradiol/farmacología , Hormona Luteinizante/sangre , Ovinos/sangre , Animales , Castración , Femenino , Hormona Luteinizante/antagonistas & inhibidores , Masculino , Pregnenodionas/farmacología , Progesterona/sangre , Estaciones del Año , Tasa de Secreción/efectos de los fármacos , Conducta Sexual Animal
9.
Reprod Fertil Dev ; 16(7): 709-15, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15740694

RESUMEN

Conversion of testosterone to oestradiol plays a major role in the feedback inhibition of gonadotrophin secretion in male sheep but little is known of the distribution or control of aromatase activity among central and peripheral tissues. Changes in activity at those sites may mediate alterations in the effectiveness of negative feedback following, for example, a change in nutrition. Using a tritiated-water assay, we quantified aromatase in several tissues in mature male sheep, assessed their contribution to oestradiol production, and tested whether activity at each site was affected by a nutritional treatment that stimulates gonadotrophin secretion. Among the brain tissues, the preoptic area had the highest concentration of activity, followed by the hypothalamus, amygdala and cortex. Among the peripheral tissues, liver and testis had the highest activity and, due to their mass, they are the major sources of circulating oestradiol. Pituitary, muscle, kidney and adipose tissues had very low aromatase levels. The nutritional stimulus increased activity in testis but not in liver or brain. We conclude that changes in aromatase activity do not mediate the effects of nutrition on steroid feedback, but aromatisation in testis, liver and brain is important in the endocrine regulation of reproduction in the mature ram.


Asunto(s)
Fenómenos Fisiológicos Nutricionales de los Animales , Aromatasa/análisis , Encéfalo/enzimología , Animales , Estradiol/metabolismo , Retroalimentación Fisiológica , Hígado/enzimología , Masculino , Ovinos , Testículo/enzimología , Distribución Tisular
10.
Horm Behav ; 44(2): 146-51, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-13129487

RESUMEN

The present study was designed to test the hypothesis that aromatization is involved in the maintenance by testosterone of the appetitive component of male sexual behavior. We measured appetitive sexual behavior by administering behavioral tests in bilevel chambers and quantifying anticipatory level changes during a 5-min period prior to introduction of a stimulus female. In addition, we recorded standard measures of consummatory male sexual behavior after the female was introduced. Following 3 weekly tests, level-changing behavior reached a plateau and remained stable for up to 10 weeks. After 10 bilevel tests, rats were given subcutaneous testosterone capsules to clamp circulating androgen at physiological levels. Rats were tested and divided into two groups that were matched for measures of sexual behavior. One group was then treated with the nonsteroidal aromatase inhibitor, Fadrozole (2.5 mg/kg/day), given subcutaneously in beta-cyclodextrin and the other group was treated with vehicle. Within 1 week of Fadrozole treatment, the number of anticipatory levels changes was significantly reduced, but not the latency to begin searching. Fadrozole treatment also significantly reduced all measures of copulatory behavior over the period of treatment and increased latencies to first mount, intromission, and ejaculation. After 8 weeks, both treatment groups were given an additional Silastic capsule filled with estradiol and tested for 4 additional weeks. Estrogen treatment partially restored level-changing behavior, mounts, and intromissions but had little effect on ejaculations. These results support the view that aromatization is important for maintaining both the appetitive and the consummatory aspects of sexual behavior in male rats.


Asunto(s)
Inhibidores de la Aromatasa , Inhibidores Enzimáticos/farmacología , Fadrozol/farmacología , Conducta Sexual Animal/efectos de los fármacos , Animales , Conducta Apetitiva/efectos de los fármacos , Conducta Apetitiva/fisiología , Conducta Consumatoria/efectos de los fármacos , Conducta Consumatoria/fisiología , Estradiol/sangre , Masculino , Motivación , Ratas , Ratas Sprague-Dawley , Conducta Sexual Animal/fisiología , Testosterona/sangre
11.
J Comp Neurol ; 439(2): 208-23, 2001 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-11596049

RESUMEN

This study mapped the regional locations of cells expressing cytochrome P450 aromatase (P450AROM) and androgen receptor (AR) mRNAs in the adult male macaque hypothalamus and amygdala by in situ hybridization histochemistry using monkey-specific cRNA probes. High densities of P450AROM and AR mRNA-containing neurons were observed in discrete hypothalamic areas involved in the regulation of gonadotropin secretion and reproductive behavior. P450AROM mRNA-containing neurons were most abundant in the medial preoptic nucleus, bed nucleus of the stria terminalis, and anterior hypothalamic area, whereas AR mRNA-containing neurons were most numerous in the ventromedial nucleus, arcuate nucleus, and tuberomamillary nucleus. Moderate to heavily labeled P450AROM mRNA-containing cells were present in the cortical and medial amygdaloid nuclei, which are known to have strong reciprocal inputs with the hypothalamus. Heavily labeled P450AROM mRNA-containing cells were found in the accessory basal amygdala nucleus, which projects to the cingulate cortex and hippocampus, areas that are important in the expression of emotional behaviors and memory processing. In contrast to P450AROM, the highest density of AR mRNA labeling in the temporal lobe was associated with the cortical amygdaloid nucleus and the pyramidal cells of the hippocampus. All areas that contained P450AROM mRNA-expressing cells also contained AR mRNA-expressing cells, but there were areas in which AR mRNA was expressed but not P450AROM mRNA. The apparent relative differences in the expression of P450AROM and AR mRNA-containing neurons within the monkey brain suggests that T acts through different signaling pathways in specific brain areas or within different cells from the same region.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Aromatasa/genética , Hipotálamo/metabolismo , Macaca fascicularis/metabolismo , ARN Mensajero/metabolismo , Receptores Androgénicos/genética , Reproducción/fisiología , Amígdala del Cerebelo/citología , Animales , Secuencia de Bases , Bioensayo , ADN Complementario/química , Hipotálamo/citología , Hibridación in Situ , Macaca fascicularis/anatomía & histología , Masculino , Datos de Secuencia Molecular , Neuronas/citología , Neuronas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ribonucleasas/análisis , Núcleos Septales/citología , Núcleos Septales/metabolismo , Homología de Secuencia de Ácido Nucleico , Factores Sexuales
12.
J Steroid Biochem Mol Biol ; 79(1-5): 247-53, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11850231

RESUMEN

In adult male primates, estrogens play a role in both gonadotropin feedback and sexual behavior. Inhibition of aromatization in intact male monkeys acutely elevates serum levels of luteinizing hormone, an effect mediated, at least partially, within the brain. High levels of aromatase (CYP19) are present in the monkey brain and regulated by androgens in regions thought to be involved in the central regulation of reproduction. Androgens regulate aromatase pretranslationally and androgen receptor activation is correlated with the induction of aromatase activity. Aromatase and androgen receptor mRNAs display both unique and overlapping distributions within the hypothalamus and limbic system suggesting that androgens and androgen-derived estrogens regulate complimentary and interacting genes within many neural networks. Long-term castrated monkeys, like men, exhibit an estrogen-dependent neural deficit that could be an underlying cause of the insensitivity to testosterone that develops in states of chronic androgen deficiency. Future studies of in situ estrogen formation in brain in the primate model are important for understanding the importance of aromatase not only for reproduction, but also for neural functions such as memory and cognition that appear to be modulated by estrogens.


Asunto(s)
Aromatasa/metabolismo , Encéfalo/enzimología , Andrógenos/metabolismo , Animales , Aromatasa/genética , Encéfalo/fisiología , Resistencia a Medicamentos , Estrógenos/metabolismo , Retroalimentación , Regulación Enzimológica de la Expresión Génica , Humanos , Hormona Luteinizante/metabolismo , Macaca mulatta , Masculino , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Conducta Sexual Animal/fisiología , Testosterona/metabolismo , Testosterona/farmacología , Distribución Tisular
13.
Brain Res ; 880(1-2): 92-101, 2000 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-11032993

RESUMEN

It is well known that certain actions of androgen are mediated through in situ aromatization to estrogen in neural target tissues. This study was undertaken to investigate androgen utilization in the hypothalamus of the female frog, Rana esculenta, through a quantification of estrogen receptors and aromatase activity during the reproductive cycle. 3H-estradiol-binding molecules were present in both the cytosol and the nuclear extract of the hypothalamus. These molecules bound specifically 3H-estradiol with high affinity (Kd 10(-10) M) and low capacity (cytosol: 1.2+/-0.4 fmol/mg protein; nuclear extract: 7.9+/-0.6 fmol/mg protein). Aromatase activity was detected in the microsomal fraction of the hypothalamus using a sensitive in vitro radiometric assay. Both aromatase activity and nuclear estrogen receptor binding fluctuated in synchrony throughout the reproductive cycle. Western blot analysis of aromatase protein revealed one immunoreactive band with a molecular weight of approximately 56 kDa. In contrast to aromatase enzyme activity, the relative levels of aromatase protein changed little during the reproductive cycle suggesting that post-translational mechanisms may be involved in regulating estrogen synthesis in the frog brain. A possible role for estrogens in the modulation of the reproductive behavior in this species is suggested.


Asunto(s)
Aromatasa/metabolismo , Hipotálamo/fisiología , Receptores de Estrógenos/metabolismo , Reproducción/fisiología , Animales , Unión Competitiva , Núcleo Celular/metabolismo , Citosol/metabolismo , Deuterio , Estradiol/metabolismo , Femenino , Cinética , Microsomas/metabolismo , Ensayo de Unión Radioligante , Rana esculenta , Estaciones del Año , Sensibilidad y Especificidad
14.
Biol Reprod ; 63(3): 872-8, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10952934

RESUMEN

Testosterone at physiological levels cannot exert negative feedback action on LH secretion in long-term castrated male monkeys. The cellular basis of this refractoriness is unknown. To study it, we compared two groups of male rhesus macaques: one group (group 1, n = 4) was castrated and immediately treated with testosterone for 30 days; the second group (group 2, n = 4) was castrated and treated with testosterone for 9 days beginning 21 days after castration. Feedback control of LH by testosterone in group 1 was normal, whereas insensitivity to its action was found in group 2. Using the endpoints of concentrations of aromatase activity (P450(AROM) messenger RNA [mRNA]) and androgen receptor mRNA in the medial preoptic anterior hypothalamus and in the medial basal hypothalamus, we found that aromatase activity in both of these tissues was significantly lower, P: < 0.01, in group 2 compared with group 1 males. P450(AROM) mRNA and androgen receptor mRNA did not differ, however. Our data suggest that the cellular basis of testosterone insensitivity after long-term castration may reside in the reduced capacity of specific brain areas to aromatize testosterone. Because P450(AROM) mRNA did not change in group 2 males, we hypothesize that an estrogen-dependent neural deficit, not involving the regulation of the P450(AROM) mRNA, occurs in long-term castrated monkeys.


Asunto(s)
Hormona Luteinizante/metabolismo , Orquiectomía , Testosterona/farmacología , Androstenodiona/sangre , Animales , Aromatasa/genética , Aromatasa/metabolismo , Ritmo Circadiano , Dihidrotestosterona/sangre , Estradiol/sangre , Retroalimentación , Hipotálamo Anterior/química , Hipotálamo Medio/química , Hormona Luteinizante/sangre , Macaca mulatta , Masculino , Área Preóptica/química , ARN Mensajero/análisis , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Testosterona/sangre , Testosterona/metabolismo
15.
Biol Reprod ; 62(6): 1818-22, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10819787

RESUMEN

We demonstrated previously that testosterone regulates aromatase activity in the anterior/dorsolateral hypothalamus of male rhesus macaques. To determine the level of the androgen effect, we developed a ribonuclease protection assay to study the effects of testosterone or dihydrotestosterone (DHT) on aromatase (P450(AROM)) mRNA in selected brain areas. Adult male rhesus monkeys were treated with testosterone or DHT. Steroids in serum were quantified by RIA. Fourteen brain regions were analyzed for P450(AROM) mRNA. Significant elevations of its message over controls (P<0.05) were found in the medial preoptic area/anterior hypothalamus of both androgen treatment groups and the medial basal hypothalamus of the testosterone-treated males. Other brain areas were not affected by androgen treatment. We conclude that testosterone and DHT regulate P450(AROM) mRNA in brain regions that mediate reproductive behaviors and gonadotropin release. The P450(AROM) mRNA of other brain areas is not androgen dependent. Brain-derived estrogens may also be important for maintaining neural circuitry in brain areas not related to reproduction. The control of P450(AROM) mRNA in these areas may differ from what we report here, but it is equally important to understand the function of in situ estrogen formation in these areas.


Asunto(s)
Andrógenos/farmacología , Aromatasa/genética , Encéfalo/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , ARN Mensajero/metabolismo , Animales , Dihidrotestosterona/sangre , Dihidrotestosterona/farmacología , Estradiol/sangre , Hipotálamo Anterior/metabolismo , Hipotálamo Medio/metabolismo , Macaca mulatta , Masculino , Área Preóptica/metabolismo , Testosterona/sangre , Testosterona/farmacología
16.
Am J Physiol ; 276(5): R1346-50, 1999 05.
Artículo en Inglés | MEDLINE | ID: mdl-10233026

RESUMEN

Testosterone and estradiol act synergistically to stimulate male sexual behavior. Previous studies demonstrated that testosterone's actions are mediated genomically. Attempts to show that estradiol acts in a similar fashion have been inconclusive. However, estrogens have been shown to exert short-latency effects by acting directly on neuronal membranes. The present experiment examined whether testosterone or estradiol rapidly facilitates copulatory behaviors in castrated sexually experienced rats. Within 35 min of administration, estradiol stimulated chemoinvestigation and frequency of mounting and reduced mount latency in a dose-dependent manner. In contrast, acute administration of testosterone did not alter sexual activity. These data demonstrate for the first time that estradiol exerts short-latency effects on copulatory behavior, providing indirect evidence that this action is mediated through a nontranscriptional mechanism.


Asunto(s)
Copulación/efectos de los fármacos , Estradiol/farmacología , Orquiectomía , Animales , Antineoplásicos Hormonales/sangre , Antineoplásicos Hormonales/farmacología , Relación Dosis-Respuesta a Droga , Estradiol/sangre , Femenino , Masculino , Ratas , Ratas Sprague-Dawley , Tiempo de Reacción/efectos de los fármacos , Tiempo de Reacción/fisiología , Testosterona/sangre , Testosterona/farmacología
17.
Neuroendocrinology ; 69(4): 290-8, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10207281

RESUMEN

The present experiment was performed to test the hypothesis that gender differences in the capacity for brain estrogen synthesis could constitute a sexually dimorphic mechanism that limits the activational effects of testosterone (T) in females, and enhances them in males. We determined the effects of treatments with equivalent levels of either T or estradiol (E2) on olfactory behavior and mounting in age-matched heterosexually naive gonadectomized male and female rats that were genitally ansthetized with lidocaine paste in order to minimize the contribution of sexually dimorphic somatosensory inputs to the expression of copulatory behavior. We found that T stimulated mounting to a greater extent in males than in females, but had equivalent effects on mount latency and genital investigation in the two sexes. On the other hand, E2 stimulated equivalent levels of mounting in males and females and reduced mount latency to a similar extent in males and females. However, E2 had a pronounced effect on the levels of genital investigation in males but not in females. Serum steroid levels and the levels of nuclear steroid receptor occupation in the brain were not different between males and females, suggesting that the behavioral differences between males and females cannot be attributed to differences in peripheral steroid metabolism or brain uptake. The results obtained corroborate previous studies suggesting that female rats normally undergo considerable male-typical behavioral masculinization during fetal development. However, such male-typical features of normal development in female rats do not extend to the regulation of preoptic aromatase activity or to the capacity of females to display olfactory behaviors in response to adult E2 exposure, functions which are sexually dimorphic even in the rat. The present results support the view that gender differences in the capacity for brain estrogen synthesis contribute to the sexually dimorphic display of T-stimulated male-typical sexual motivation and copulatory behavior in rats.


Asunto(s)
Copulación/efectos de los fármacos , Estradiol/farmacología , Caracteres Sexuales , Conducta Sexual Animal/efectos de los fármacos , Testosterona/farmacología , Animales , Núcleo Celular/metabolismo , Femenino , Hipotálamo/ultraestructura , Masculino , Orquiectomía , Ovariectomía , Ratas , Ratas Sprague-Dawley , Receptores Androgénicos/metabolismo , Receptores de Estrógenos/metabolismo , Olfato
18.
Brain Res ; 792(2): 271-6, 1998 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-9593936

RESUMEN

The level of aromatase in the preoptic area of rats is transcriptionally regulated through a specific androgen-receptor mediated mechanism and can be used as a measure of central androgenic effect. Therefore, several commonly abused anabolic-androgenic steroids (AAS) were tested for their ability to induce aromatase activity in the preoptic area of castrated rats. In addition, we determined the relative binding affinities of these compounds for the androgen receptor, as well as their ability to bind androgen receptor in vivo following subcutaneous injections. All of the AAS compounds tested significantly stimulated POA aromatase activity above castrate levels. The compounds that produced the greatest stimulation of aromatase activity were those that bound most avidly to the androgen receptor in vitro (i.e., testosterone, dihydrotestosterone and nandrolone). In contrast, the 17alpha-alkylated compounds that were tested (stanozolol, danazol, methandrostenolone) modestly stimulated aromatase and were weak competitors for the androgen receptor. The subcutaneous injection of AAS compounds increased the concentrations of occupied nuclear androgen receptors in the brain, but the magnitude of effect was not related to their potency for inducing aromatase or their relative binding affinity for the androgen receptor suggesting that androgen receptor occupancy in POA is not correlated with the action of androgen on aromatase. The present results help explain the behavioral effects of AAS compounds in rats.


Asunto(s)
Anabolizantes/farmacología , Andrógenos , Andrógenos/farmacología , Aromatasa/metabolismo , Área Preóptica/enzimología , Anabolizantes/metabolismo , Andrógenos/metabolismo , Animales , Antineoplásicos Hormonales/farmacología , Danazol/farmacología , Dihidrotestosterona/farmacología , Activación Enzimática/efectos de los fármacos , Antagonistas de Estrógenos/farmacología , Inyecciones Subcutáneas , Masculino , Metandrostenolona/farmacología , Nandrolona/farmacología , Orquiectomía , Tamaño de los Órganos , Área Preóptica/química , Próstata/anatomía & histología , Próstata/química , Próstata/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores Androgénicos/metabolismo , Estanozolol/farmacología , Testosterona/farmacología
19.
J Steroid Biochem Mol Biol ; 61(3-6): 365-74, 1997 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9365212

RESUMEN

The basis of functional gender differences in adult responsiveness to testosterone (T) is not yet understood. Conversion of T to estradiol by cytochrome P450 aromatase in the medial preoptic area is required for the full expression of male sexual behavior in rats. High levels of aromatase are found in the medial preoptic nucleus (MPN) and in an interconnected group of sexually dimorphic nuclei which mediate masculine sexual behavior. Within this neural circuit, aromatase is regulated by T, acting through an androgen receptor (AR)-mediated mechanism. This arrangement constitutes a feedforward system because T is both the regulator and the major substrate of aromatase. Preoptic aromatase is thus more active in adult males than in females because of normal sex differences in circulating androgen levels. However, the mechanism of enzyme induction also appears to be sexually dimorphic because equivalent physiological doses of T stimulate aromatase to a greater extent in males than in females. Dose-response studies indicate that the sex difference is apparent over a range of circulating T concentrations and constitute a gender difference in T efficacy, but not potency. Sex differences in aromatase correlate with sex differences in nuclear AR concentrations in most regions of the sexually dimorphic neural circuit, but not in MPN. These results suggest that males may have larger populations of target cells in which aromatase is regulated by androgen, but the lack of a gender difference in AR levels in the MPN suggests that differences in post-receptor mechanisms could also be involved. Measurements of aromatase mRNA in androgen-treated gonadectomized rats demonstrate that sex difference in regulation is exerted pretranslationally. Taken together these results demonstrate a sexually dimorphic mechanism that could potentially limit the action of T in females, and may relate to the enhanced expression of T-stimulated sexual behaviors in males.


Asunto(s)
Andrógenos/fisiología , Aromatasa/fisiología , Encéfalo/fisiología , Regulación Enzimológica de la Expresión Génica , Animales , Femenino , Masculino , Ratas , Factores Sexuales
20.
Brain Res Bull ; 44(4): 351-7, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9370199

RESUMEN

Brain aromatase plays an important role in the regulation of adult reproductive behavior in male rodents. This report focuses on recent experiments from our laboratory that examined the distribution and regulation of aromatase mRNA in the rat brain. Aromatase mRNA was measured by a highly sensitive ribonuclease protection assay using a 32P-labeled antisense RNA probe that was complimentary to the 5' coding region of rat aromatase mRNA. This probe protects two RNA fragments in rat brain tissue: a 430-nt length fragment and a shorter 300-nt fragment. The presence of the 300-nt RNA fragment is not associated with enzyme activity in the rat brain and appears to represent an alternative brain-specific aromatase transcript whose function, if any, is unknown. In contrast, the 430-nt RNA fragment represents mRNA, which is thought to encode functional aromatase enzyme because its levels are correlated with aromatase activity concentrations in preoptic area, hypothalamus, amygdala, and ovary. Aromatase activity and mRNA levels in the preoptic area and hypothalamus decreased by 7 days after castration and were maintained at intact levels by treatment with testosterone and dihyhdrotestosterone, but not with estradiol. In contrast, neither aromatase activity nor mRNA levels in the amygdala are affected by castration or hormone replacement. In addition, sex differences in the regulation of aromatase mRNA were apparent in both the preoptic area and hypothalamus. These results demonstrate that androgens regulate the transcription or stability of aromatase mRNA in specific brain areas. Moreover, they suggest that gender differences in androgen responsiveness play an important role in regulating gene expression in the adult rat brain.


Asunto(s)
Aromatasa/biosíntesis , Encéfalo/enzimología , Regulación Enzimológica de la Expresión Génica , Transcripción Genética , Empalme Alternativo , Animales , Dihidrotestosterona/farmacología , Estradiol/farmacología , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Masculino , Orquiectomía , Especificidad de Órganos , Ovario/enzimología , Área Preóptica/enzimología , Sondas ARN , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Testosterona/farmacología , Transcripción Genética/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA