Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Horm Behav ; 146: 105279, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36370679

RESUMEN

Androgen receptors (AR) are crucial in the control of male sexual behavior and sex preference. AR are particularly concentrated in areas related with the neuroendocrine control of sex preference including the medial amygdala (MeA), the ventromedial nucleus of the hypothalamus (VMH), the bed nucleus of the stria terminalis (BNST), the medial preoptic area (MPOA), the nucleus accumbens (Acb), the suprachiasmatic (SCh) and supraoptic (SO) nuclei, but also seem to be important for the control of reproductive processes in the hippocampus (CA1-CA4 and dentate gyrus, DG). In the present study we analyzed the density of AR in these brain areas of adult male rats with sexual preference (established in a three-compartment box). Same-sex preference was produced in male rats by the prenatal administration of the aromatase inhibitor, letrozole (0.56 µg/kg/ml s.c. G10-22) that usually produces 1-2 animals per litter with same sex preference, while the others retain a female sex preference. We also included a group of proestrus females that had a clear preference for a sexually active male. AR were analyzed by immunocytochemistry using PG21 as primary antibody. We also measured total plasma testosterone concentrations by radioimmunoassay. In males with same sex preference there was a specific AR overexpression in CA3 and CA4 that suggests a feminized pattern because females in proestrus trend to show a higher density of AR in these hippocampal areas. Sex differences in AR density were found in the anterior cingulate cortex (ACg) and frontoparietal cortex (FrPa). Serum levels of testosterone did not differ between groups. Data are discussed based on the role of AR in the hippocampus.


Asunto(s)
Receptores Androgénicos , Núcleos Septales , Animales , Femenino , Masculino , Ratas , Receptores Androgénicos/metabolismo , Núcleos Septales/metabolismo , Encéfalo/metabolismo , Testosterona , Conducta Sexual
2.
Am J Physiol Heart Circ Physiol ; 315(5): H1393-H1401, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30095996

RESUMEN

Polycystic ovary syndrome is a complex and common disorder in women, and those affected experience an increased burden of cardiovascular disease. It is an intergenerational syndrome, as affected women with high androgen levels during pregnancy "program" fetal development, leading to a similar phenotype in their female offspring. The effect of excess maternal testosterone exposure on fetal cardiomyocyte growth and maturation is unknown. Pregnant ewes received biweekly injections of vehicle (control) or 100 mg testosterone propionate between 30 and 59 days of gestation (early T) or between 60 and 90 days of gestation (late T). Fetuses were delivered at ~135 days of gestation, and their hearts were enzymatically dissociated to measure cardiomyocyte growth (dimensional measurements), maturation (proportion binucleate), and proliferation (nuclear Ki-67 protein). Early T depressed serum insulin-like growth factor 1 and caused intrauterine growth restriction (IUGR; P < 0.0005). Hearts were smaller with early T ( P < 0.001) due to reduced cardiac myocyte maturation ( P < 0.0005) and proliferation ( P = 0.017). Maturation was also lower in male than female fetuses ( P = 0.004) independent of treatment. Late T did not affect cardiac growth. Early excess maternal testosterone exposure depresses circulating insulin-like growth factor 1 near term and causes IUGR in both female and male offspring. These fetuses have small, immature hearts with reduced proliferation, which may reduce cardiac myocyte endowment and predispose to adverse cardiac growth in postnatal life. While excess maternal testosterone exposure leads to polycystic ovary syndrome and cardiovascular disease in female offspring, it may also predispose to complications of IUGR and cardiovascular disease in male offspring. NEW & NOTEWORTHY Using measurements of cardiac myocyte growth and maturation in an ovine model of polycystic ovary syndrome, this study demonstrates that early gestation excess maternal testosterone exposure reduces near-term cardiomyocyte proliferation and maturation in intrauterine growth-restricted female and male fetuses. The effect of testosterone is restricted to exposure during a specific period early in pregnancy, and the effects appear mediated through reduced insulin-like growth factor 1 signaling. Furthermore, male fetuses, regardless of treatment, had fewer mature cardiomyocytes than female fetuses.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Retardo del Crecimiento Fetal/patología , Corazón Fetal/patología , Miocitos Cardíacos/patología , Propionato de Testosterona , Animales , Modelos Animales de Enfermedad , Femenino , Retardo del Crecimiento Fetal/sangre , Retardo del Crecimiento Fetal/inducido químicamente , Corazón Fetal/metabolismo , Edad Gestacional , Factor I del Crecimiento Similar a la Insulina/metabolismo , Antígeno Ki-67/metabolismo , Masculino , Exposición Materna , Miocitos Cardíacos/metabolismo , Embarazo , Factores Sexuales , Oveja Doméstica
4.
Neuroendocrinology ; 97(3): 271-82, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23051877

RESUMEN

BACKGROUND: Men have a higher stroke incidence compared to women until advanced age. The contribution of hormones to these sex differences has been extensively debated. In experimental stroke, estradiol is neuroprotective, whereas androgens are detrimental. However, prior studies have only examined the effects of acute treatment paradigms; therefore, the timing and mechanism by which ischemic sexual dimorphism arises are unknown. METHODS: The effects of exogenous neonatal androgen exposure on subsequent injury induced by middle cerebral artery occlusion in adulthood in male rats were examined. Rats were administered vehicle (oil), testosterone propionate (TP) or the non-aromatizable androgen dihydrotestosterone (DHT) for 5 days after birth. At 3 months of age, a focal stroke was induced. RESULTS: Testosterone-treated rats (but not DHT-treated animals) had decreased infarct volumes (20 vs. 33%, p < 0.05) as well as increased estradiol levels (39.4 vs. 18.6 pg/ml, p < 0.0001) compared to oil-treated animals. TP-injected males had increased testicular aromatase (P450arom) levels (3.6 vs. 0.2 ng/ml, p < 0.0001) compared to oil-treated males. The level of X-linked inhibitor of apoptosis, the primary endogenous inhibitor of caspase-induced apoptosis, was increased in TP-treated rats compared with the oil-treated males. CONCLUSIONS: Neonatal exposure to exogenous testosterone upregulates testicular aromatase expression in male rats and leads to adult neuroprotection secondary to changes in serum estradiol levels and cell death proteins. This study suggests that early exposure to gonadal hormones can have dramatic effects on the response to adult cerebrovascular injury.


Asunto(s)
Accidente Cerebrovascular/prevención & control , Propionato de Testosterona/farmacología , Animales , Animales Recién Nacidos , Aromatasa/metabolismo , Infarto Encefálico/metabolismo , Infarto Encefálico/patología , Infarto Encefálico/prevención & control , Dihidrotestosterona/administración & dosificación , Dihidrotestosterona/farmacología , Estradiol/sangre , Infarto de la Arteria Cerebral Media , Masculino , Ratas , Accidente Cerebrovascular/sangre , Accidente Cerebrovascular/patología , Testículo/metabolismo , Propionato de Testosterona/administración & dosificación , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo
5.
Endocrinology ; 164(10)2023 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-37610243

RESUMEN

We previously demonstrated that treating fetal lambs on gestational day 62 with the long-acting gonadotrophin-releasing hormone (GnRH) antagonist degarelix (DG) suppresses pituitary-testicular function during midgestation. The objective of this study was to investigate whether impaired gonadotrophic drive during this fetal period has enduring effects on sexual differentiation and reproductive function in adult male sheep. We assessed the effects of prenatal administration of DG, with or without testosterone (T) replacement, on various sexually dimorphic behavioral traits in adult rams, including sexual partner preferences, as well as neuroendocrine responsiveness and testicular function. Our findings revealed that DG treatment had no effect on genital differentiation or somatic growth. There were some indications that DG treatment suppressed juvenile play behavior and adult sexual motivation; however, male-typical sexual differentiation of reproductive behavior, sexual partner preference, and gonadotropin feedback remained unaffected and appeared to be fully masculinized and defeminized. DG-treated rams showed an increased LH response to GnRH stimulation and a decreased T response to human chorionic gonadotropin stimulation, suggesting impaired Leydig cell function and reduced T feedback. Both effects were reversed by cotreatment with T propionate. DG treatment also suppressed the expression of CYP17 messenger RNA, a key enzyme for T biosynthesis. Despite the mild hypogonadism induced by DG treatment, ejaculate volume, sperm motility, and sperm morphology were not affected. In summary, these results suggest that blocking GnRH during midgestation does not have enduring effects on brain sexual differentiation but does negatively affect the testes' capacity to synthesize T.


Asunto(s)
Enfermedades de la Hipófisis , Testículo , Adulto , Humanos , Femenino , Embarazo , Masculino , Ovinos , Animales , Diferenciación Sexual , Semen , Motilidad Espermática , Encéfalo , Oveja Doméstica , Hormona Liberadora de Gonadotropina
6.
Front Neuroendocrinol ; 32(2): 164-9, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21215767

RESUMEN

The sheep offers a unique mammalian model in which to study paradoxical same-sex sexual partner preferences. Variations in sexual partner preferences occur spontaneously with as many as 8% of rams in a population exhibiting a sexual preference for other rams (male-oriented). The current review presents an overview and update of the male-oriented ram model and discusses several theories that have been invoked to explain same-sex preferences in this species. Although our understanding of the biological determinants and underlying neural substrates of sexual attraction and mate selection are far from complete, compelling evidence is discussed that supports the idea that neural substrates regulating sexual partner preferences are organized during prenatal development. The challenge for future research will be to construct an integrated picture of how hormones, genes, and experience shape sexual partner preference.


Asunto(s)
Conducta Sexual Animal/fisiología , Oveja Doméstica/fisiología , Animales , Conducta Animal , Encéfalo/efectos de los fármacos , Conducta de Elección/fisiología , Período Crítico Psicológico , Ciclo Estral/fisiología , Femenino , Masculino , Área Preóptica/anatomía & histología , Atractivos Sexuales/farmacología , Diferenciación Sexual/efectos de los fármacos , Diferenciación Sexual/fisiología , Olfato/fisiología , Predominio Social , Testosterona/fisiología , Órgano Vomeronasal/fisiología
7.
PLoS One ; 17(5): e0263319, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35552544

RESUMEN

The sheep is a valuable model to test whether hormone mechanisms that sexually differentiate the brain underlie the expression of sexual partner preferences because as many as 8% of rams prefer same-sex partners. Epigenetic factors such as DNA methylation act as mediators in the interaction between steroid hormones and the genome. Variations in the epigenome could be important in determining morphological or behavior differences among individuals of the same species. In this study, we explored DNA methylation differences in the hypothalamus of male oriented rams (MORs) and female oriented rams (FORs). We employed reduced representation bisulfite sequencing (RRBS) to generate a genome-wide map of DNA methylation and RNA-Seq to profile the transcriptome. We found substantial DNA methylation and gene expression differences between FORs and MORs. Although none of the differentially methylated genes yielded significant functional terms directly associated with sex development, three differentially expressed genes were identified that have been associated previously with sexual behaviors. We hypothesize that these differences are involved in the phenotypic variation in ram sexual partner preferences, whereas future studies will have to find the specific mechanisms. Our results add an intriguing new dimension to sheep behavior that should be useful for further understanding epigenetic and transcriptomic involvement.


Asunto(s)
Conducta Sexual Animal , Parejas Sexuales , Animales , Metilación de ADN , Femenino , Expresión Génica , Humanos , Hipotálamo , Masculino , Ovinos/genética , Oveja Doméstica
8.
Endocrinology ; 163(2)2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34958103

RESUMEN

The specific role of gonadotropin-releasing hormone (GnRH) on brain sexual differentiation remains unclear. To investigate whether gonadotropin and, in turn, testosterone (T) secretion is regulated by GnRH during the critical period for brain differentiation in sheep fetuses, we attempted to selectively suppress pituitary-testicular activation during midgestation with the long-acting GnRH antagonist degarelix. Fetuses received subcutaneous injections of the antagonist or vehicle on day 62 of gestation. After 2 to 3 weeks we examined consequences of the intervention on baseline and GnRH-stimulated plasma luteinizing hormone (LH) and T levels. In addition, we measured the effect of degarelix-treatment on messenger RNA (mRNA) expression for the pituitary gonadotropins and key gonadal steroidogenic enzymes. Baseline and GnRH-stimulated plasma LH levels were significantly suppressed in degarelix-treated male and female fetuses compared to control values. Similarly, T concentrations were suppressed in degarelix-treated males. The percentage of LHß-immunoreactive cells colocalizing c-fos was significantly reduced by degarelix treatment indicating that pituitary sensitivity was inhibited. Degarelix treatment also led to the significant suppression of mRNA expression coding for the pituitary gonadotropin subunits and for the gonadal enzymes involved in androgen synthesis. These findings demonstrate that pharmacologic inhibition of GnRH early in gestation results in suppression of LH secretion and deficits in the plasma T levels of male lamb fetuses. We conclude that GnRH signaling plays a pivotal role for regulating T exposure during the critical period of sheep gestation when the brain is masculinized. Thus, disturbance to gonadotropin secretion during this phase of gestation could have long-term consequence on adult sexual behaviors and fertility.


Asunto(s)
Edad Gestacional , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Gonadotropinas Hipofisarias/metabolismo , Oligopéptidos/administración & dosificación , Adenohipófisis/embriología , Ovinos/embriología , Animales , Encéfalo/embriología , Femenino , Sangre Fetal/química , Hormona Liberadora de Gonadotropina/administración & dosificación , Hormona Liberadora de Gonadotropina/fisiología , Gonadotropinas Hipofisarias/genética , Inyecciones Subcutáneas/veterinaria , Hormona Luteinizante/sangre , Masculino , Ovario/química , Ovario/embriología , Adenohipófisis/química , Adenohipófisis/efectos de los fármacos , Embarazo , ARN Mensajero/análisis , Diferenciación Sexual/fisiología , Testículo/química , Testículo/embriología , Testosterona/sangre
9.
Neurosci Biobehav Rev ; 114: 12-15, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32311371

RESUMEN

The sheep is a valuable model to test whether hormone mechanisms that sexually differentiate the brain underlie the expression of sexual partner preferences because as many as 8% of rams prefer same-sex partners. This review presents an overview and update of the experimental evidence that supports this hypothesis. New evidence is presented that demonstrates a critical role for kisspeptin-GnRH signaling for regulating stable fetal testosterone levels necessary for masculinization of brain and behavior. Although these studies provide substantial support for the idea that prenatal hormones program sexual preferences, further experimentation is needed to establish causality.


Asunto(s)
Conducta Sexual Animal , Testosterona , Animales , Biología , Encéfalo , Femenino , Embarazo , Ovinos
10.
Endocrinology ; 161(4)2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32005991

RESUMEN

Evidence suggests that the hypothalamic-pituitary-gonadal (HPG) axis is active during the critical period for sexual differentiation of the ovine sexually dimorphic nucleus, which occurs between gestational day (GD) 60 and 90. Two possible neuropeptides that could activate the fetal HPG axis are kisspeptin and neurokinin B (NKB). We used GD85 fetal lambs to determine whether intravenous administration of kisspeptin-10 (KP-10) or senktide (NKB agonist) could elicit luteinizing hormone (LH) release. Immunohistochemistry and fluorescent in situ hybridization (FISH) were employed to localize these peptides in brains of GD60 and GD85 lamb fetuses. In anesthetized fetuses, KP-10 elicited robust release of LH that was accompanied by a delayed rise in serum testosterone in males. Pretreatment with the GnRH receptor antagonist (acyline) abolished the LH response to KP-10, confirming a hypothalamic site of action. In unanesthetized fetuses, senktide, as well as KP-10, elicited LH release. The senktide response of females was greater than that of males, indicating a difference in NKB sensitivity between sexes. Gonadotropin-releasing hormone also induced a greater LH discharge in females than in males, indicating that testosterone negative feedback is mediated through pituitary gonadotrophs. Kisspeptin and NKB immunoreactive cells in the arcuate nucleus were more abundant in females than in males. Greater than 85% of arcuate kisspeptin cells costained for NKB. FISH revealed that the majority of these were kisspeptin/NKB/dynorphin (KNDy) neurons. These results support the hypothesis that kisspeptin-GnRH signaling regulates the reproductive axis of the ovine fetus during the prenatal critical period acting to maintain a stable androgen milieu necessary for brain masculinization.


Asunto(s)
Hipotálamo/efectos de los fármacos , Kisspeptinas/farmacología , Hormona Luteinizante/sangre , Testosterona/sangre , Animales , Femenino , Feto , Hormona Liberadora de Gonadotropina/farmacología , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Masculino , Neuroquinina B/metabolismo , Oligopéptidos/farmacología , Fragmentos de Péptidos/farmacología , Embarazo , Receptores de Kisspeptina-1/agonistas , Receptores de Neuroquinina-3/agonistas , Ovinos , Sustancia P/análogos & derivados , Sustancia P/farmacología
11.
Int J Oncol ; 34(5): 1319-27, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19360344

RESUMEN

Expression and activity of CC motif ligand 2 (CCL2) is down-regulated by curcumin, the active phytochemical ingredient of turmeric (Curcuma longa), a dietary supplement often self-prescribed to promote prostate health. CCL2 is a potent chemotactic factor of prostate cancer (PCa) with important roles in development of bone metastasis. The relationship between CCL2 and curcumin, however, has not been studied in PCa. Adhesion, invasion and motility of PC-3 cells were measured in response to exposure to curcumin (30 microM; 18 h), CCL2 (100 ng/ml; 18 h) or PMA (100 ng/ml; 18 h). CCL2 mRNA expression and protein secretion levels were measured by real-time PCR and ELISA respectively. Curcumin significantly blocked CCL2 induced adhesion, invasion and motility. Curcumin also significantly suppressed the mRNA expression and secreted CCL2 protein levels. The addition of PMA, a protein kinase C (PKC) activator, blocked the effects of curcumin, leading to an increase in CCL2 expression as well as an increase in PC-3 cell adhesion, invasion and motility. The introduction of a PKC inhibitor, however, blocked the effects of CCL2. We also found that curcumin, CCL2 and PMA, in part, function through the differential regulation of the proteolytic protein matrix metalloproteinase (MMP)-9. These data indicate a potential mechanism; by which curcumin can block the chemotactic effects of CCL2 on PCa. Curcumin exerts potential anti-metastatic effects in bone-derived PCa cells by blocking CCL2 mediated actions on invasion, adhesion and motility, in part through differential regulation of PKC and MMP-9 signaling.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Curcumina/farmacología , Neoplasias de la Próstata/patología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Movimiento Celular/genética , Quimiocina CCL2/fisiología , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Evaluación Preclínica de Medicamentos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Modelos Biológicos , Invasividad Neoplásica , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Proteína Quinasa C/fisiología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Procesamiento Proteico-Postraduccional/genética , Células Tumorales Cultivadas
12.
Horm Behav ; 55(5): 611-20, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19446078

RESUMEN

The question of what causes a male animal to seek out and choose a female as opposed to another male mating partner is unresolved and remains an issue of considerable debate. The most developed biologic theory is the perinatal organizational hypothesis, which states that perinatal hormone exposure mediates sexual differentiation of the brain. Numerous animal experiments have assessed the contribution of perinatal testosterone and/or estradiol exposure to the development of a male-typical mate preference, but almost all have used hormonally manipulated animals. In contrast, variations in sexual partner preferences occur spontaneously in domestic rams, with as many as 8% of the population exhibiting a preference for same-sex mating partners (male-oriented rams). Thus, the domestic ram is an excellent experimental model to study possible links between fetal neuroendocrine programming of neural mechanisms and adult sexual partner preferences. In this review, we present an overview of sexual differentiation in relation to sexual partner preferences. We then summarize results that test the relevance of the organizational hypothesis to expression of same-sex sexual partner preferences in rams. Finally, we demonstrate that the sexual differentiation of brain and behavior in sheep does not depend critically on aromatization of testosterone to estradiol.


Asunto(s)
Andrógenos/fisiología , Encéfalo/fisiología , Preferencia en el Apareamiento Animal/fisiología , Diferenciación Sexual/fisiología , Animales , Femenino , Desarrollo Fetal/fisiología , Homosexualidad/fisiología , Masculino , Ovinos
13.
Alcohol Clin Exp Res ; 33(12): 2077-87, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19740134

RESUMEN

BACKGROUND: Allopregnanolone (ALLO) is a progesterone derivative that rapidly potentiates gamma-aminobutyric acid(A) (GABA(A)) receptor-mediated inhibition and modulates symptoms of ethanol withdrawal. Because clinical and preclinical data indicate that ALLO levels are inversely related to symptoms of withdrawal, the present studies determined whether ethanol dependence and withdrawal differentially altered plasma and cortical ALLO levels in mice selectively bred for differences in ethanol withdrawal severity and determined whether the alterations in ALLO levels corresponded to a concomitant change in activity and expression of the biosynthetic enzyme 5alpha-reductase. METHODS: Male Withdrawal Seizure-Prone (WSP) and -Resistant (WSR) mice were exposed to 72 hours ethanol vapor or air and euthanized at select times following removal from the inhalation chambers. Blood was collected for analysis of ALLO and corticosterone levels by radioimmunoassay. Dissected amygdala, hippocampus, midbrain, and cortex as well as adrenals were examined for 5alpha-reductase enzyme activity and expression levels. RESULTS: Plasma ALLO was decreased significantly only in WSP mice, and this corresponded to a decrease in adrenal 5alpha-reductase expression. Cortical ALLO was decreased up to 54% in WSP mice and up to 46% in WSR mice, with a similar decrease in cortical 5alpha-reductase activity during withdrawal in the lines. While cortical gene expression was significantly decreased during withdrawal in WSP mice, there was a 4-fold increase in expression in the WSR line during withdrawal. Hippocampal 5alpha-reductase activity and gene expression was decreased only in dependent WSP mice. CONCLUSIONS: These results suggest that there are line and brain regional differences in the regulation of the neurosteroid biosynthetic enzyme 5alpha-reductase during ethanol dependence and withdrawal. In conjunction with the finding that WSP mice exhibit reduced sensitivity to ALLO during withdrawal, the present results are consistent with the hypothesis that genetic differences in ethanol withdrawal severity are due, in part, to modulatory effects of GABAergic neurosteroids such as ALLO.


Asunto(s)
Alcoholismo/metabolismo , Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Oxidorreductasas/sangre , Pregnanolona/sangre , Síndrome de Abstinencia a Sustancias/metabolismo , Administración por Inhalación , Alcoholismo/enzimología , Animales , Encéfalo/patología , Química Encefálica/efectos de los fármacos , Depresores del Sistema Nervioso Central/administración & dosificación , Depresores del Sistema Nervioso Central/sangre , Etanol/administración & dosificación , Etanol/sangre , Regulación Enzimológica de la Expresión Génica/genética , Hipocampo/enzimología , Hidrocortisona/sangre , Masculino , Ratones , Ratones Endogámicos , Radioinmunoensayo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Especificidad de la Especie , Síndrome de Abstinencia a Sustancias/enzimología
14.
Psychoneuroendocrinology ; 109: 104387, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31465941

RESUMEN

Sex differences in the brain and behavior are produced by the perinatal action of testosterone, which is converted into estradiol by the enzyme aromatase in the brain. Although magnetic resonance imaging (MRI) has been widely used in humans to study these differences, the use of animal models, where hormonal status can be properly manipulated, is necessary to explore the mechanisms involved. We used sheep, a recognized model in the field of neuroendocrinology, to assess brain morphological and functional sex differences and their regulation by adult gonadal hormones. To this end, we performed voxel-based morphometry and a resting-state functional MRI approach to assess sex differences in gonadally intact animals. We demonstrated significant sex differences in gray matter concentration (GMC) at the level of the gonadotropic axis, i.e., not only within the hypothalamus and pituitary but also within the hippocampus and the amygdala of intact animals. We then performed the same analysis one month after gonadectomy and found that some of these differences were reduced, especially in the hypothalamus and amygdala. By contrast, we found few differences in the organization of the functional connectome between males and females either before or after gonadectomy. As a whole, our study identifies brain regions that are sexually dimorphic in the sheep brain at the resolution of the MRI and highlights the role of gonadal hormones in the maintenance of these differences.


Asunto(s)
Encéfalo/efectos de los fármacos , Hormonas Gonadales/metabolismo , Caracteres Sexuales , Amígdala del Cerebelo/metabolismo , Animales , Encéfalo/diagnóstico por imagen , Castración/métodos , Estradiol/farmacología , Femenino , Hormonas Gonadales/fisiología , Sustancia Gris/diagnóstico por imagen , Sustancia Gris/efectos de los fármacos , Hipotálamo/metabolismo , Imagen por Resonancia Magnética/métodos , Masculino , Ovinos , Testosterona/farmacología
15.
Behav Neurosci ; 122(5): 955-62, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18823152

RESUMEN

Behavioral and physiological data suggest that the striatal dopaminergic system is important in the production and execution of sequential movements. Striatal function is also modulated by sex hormones, and previous studies show that estradiol is related to sequential movement in women. The authors examined whether sex hormones are involved in the production of sequential movement in healthy older and younger men. Testosterone was modified for a 6-week period such that levels in older men matched those of younger men, the conversion of testosterone to estradiol was blocked, the production of testosterone was blocked, or the men received no treatment (placebo). Sequential movement was measured before and after hormone treatment. Older men were slower and more accurate than younger men on the sequential movement task pre- and posttreatment. Hormone manipulation had no effect on movement speed. Hormone levels were not correlated with sequential movement performance in either older or younger men, suggesting that sex hormones do not modulate sequential movement in men, and hormone replacement may not restore a loss of sequential movement ability in elderly men or men with Parkinson's disease.


Asunto(s)
Andrógenos/sangre , Estrógenos/sangre , Movimiento/fisiología , Testosterona/sangre , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Análisis de Varianza , Anastrozol , Andrógenos/administración & dosificación , Inhibidores de la Aromatasa , Hormona Liberadora de Gonadotropina/administración & dosificación , Humanos , Leuprolida/administración & dosificación , Masculino , Persona de Mediana Edad , Movimiento/efectos de los fármacos , Nitrilos/administración & dosificación , Radioinmunoensayo , Tiempo de Reacción/efectos de los fármacos , Análisis de Regresión , Testosterona/administración & dosificación , Factores de Tiempo , Triazoles/administración & dosificación
16.
Neuroendocrinology ; 87(4): 206-15, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18223310

RESUMEN

Accumulating evidence in rodents suggests that a prolactin locally synthesized and released within the brain can act together with that taken up from the circulation to modulate neuroendocrine responses. The present study was designed to identify the regional patterns of prolactin expression in the adult and developing sheep brain. Specifically, we tested the hypothesis that prolactin is expressed in regions of the adult and fetal sheep brain that are critical in the development of neuroendocrine homeostatic and behavioral functions. The expression of prolactin protein in sheep brain was demonstrated by Western blot analysis and brain prolactin mRNA was detected and sequenced using RT-PCR. In situ hybridization histochemistry revealed that prolactin mRNA was expressed in the medial preoptic area, periventricular preoptic nucleus, bed nucleus of the stria terminalis, and in the paraventricular nucleus of the hypothalamus, particularly the ventral region. The neuroanatomical distribution of prolactin mRNA was best visualized in the fetus and prolactin-immunoreactive neurons could also be identified in late gestation fetuses. Brain prolactin mRNA was expressed as early as day 60 of gestation and increased as the fetus aged and peaked at day 135 (term = 147 days). Prolactin mRNA expression did not exhibit a sex difference in the preoptic area, but in the amygdala prolactin mRNA was significantly higher in females than in males at day 100 of gestation. We conclude that prolactin expressed in adult and fetal sheep brain could be involved in neurodevelopment and/or modulation of the neuroendocrine stress axis, although it is too early to rule out other possibilities given the diverse actions that have been attributed to prolactin.


Asunto(s)
Encéfalo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Prolactina/genética , Ovinos/genética , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Secuencia de Bases , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Embrión de Mamíferos , Desarrollo Fetal/genética , Datos de Secuencia Molecular , Sistemas Neurosecretores/metabolismo , Prolactina/metabolismo , ARN Mensajero/metabolismo , Análisis de Secuencia de ADN , Ovinos/embriología , Ovinos/crecimiento & desarrollo , Estrés Psicológico/genética , Estrés Psicológico/metabolismo , Distribución Tisular
17.
Endocrinology ; 148(9): 4450-7, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17540718

RESUMEN

A sexually dimorphic nucleus that can be identified in adult sheep by its characteristic pattern of cytochrome P450 aromatase mRNA exists in the preoptic/anterior hypothalamic area and is called the ovine sexually dimorphic nucleus (oSDN). In other species, male-typical sexually dimorphic preoptic nuclei develop under the influence of gonadal testosterone exposure. Thus, we hypothesized that the oSDN develops before birth in the sheep and is organized by exposure to testosterone. To test this, we determined whether an identifiable oSDN is present in the fetal lamb brain at d 130-140 gestation (term approximately 150 d). In situ hybridization for aromatase mRNA revealed a cell group in the caudal preoptic area that corresponded to the oSDN in adults. Quantitative analysis showed that the mean volume of the oSDN in late-gestation fetuses was significantly larger in male than in female lamb fetuses. We next treated a group of pregnant ewes with testosterone propionate (TP) from d 30-90 gestation and measured oSDN volumes in TP-exposed and control fetuses on d 135 gestation. The mean volume of the oSDN in female fetuses exposed to TP was significantly larger than in control females and also larger than in control and TP-exposed males. Taken together, these data demonstrate that testosterone acts during a prenatal critical period to organize the development of aromatase-expressing neurons into the male-typical oSDN in sheep.


Asunto(s)
Área Preóptica/embriología , Diferenciación Sexual , Propionato de Testosterona/farmacología , Animales , Aromatasa/genética , Desarrollo Embrionario/efectos de los fármacos , Desarrollo Embrionario/fisiología , Femenino , Hibridación in Situ , Masculino , Embarazo , Área Preóptica/efectos de los fármacos , Ovinos
18.
J Cereb Blood Flow Metab ; 27(1): 135-41, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16736049

RESUMEN

Female animals are protected from ischemic brain damage relative to age-matched males, in part through protection provided by endogenous estradiol. In brain, estradiol is produced from testosterone by cytochrome P450 aromatase (cyp 19), a steroid synthetic enzyme present in astrocytes. We tested the hypothesis that astrocytes derived from neonatal female rat brain are less susceptible than male cells to oxygen-glucose deprivation (OGD), and that this endogenous protection is associated with enhanced aromatase activity. Primary cultured cortical astrocytes were prepared from male and female rat pups separately and grown to confluence in estrogen-free media. Cell death in response to OGD, alone or in combination with hydrogen peroxide, lipopolysaccharides, interleukin-1beta, tissue necrosis factor-alpha, or nitric oxide (NO) donor diethylenetriamine/nitric oxide adduct (DETA/NO) was quantified as the ratio of propidium iodide to calcein AM-positive cells. Aromatase activity and cyp19 mRNA and protein levels were measured in cultures from each sex. Female astrocytes are more resistant to OGD and oxidant cell death induced by H(2)O(2) , but sustain greater cell death when inflammatory mediators are combined with OGD compared with OGD alone. Media transfer from female to male cells conferred protection against OGD-induced cell death. Aromatase activity and expression is greater in female than in male astrocytes. The aromatase inhibitor, Arimidex (100 nmol/L), abolishes sex differences in OGD-induced cell death, whereas treatment with 17beta-estradiol (10 nmol/L) protects cells of either sex. We conclude that astrocytes isolated from neonatal cortex exhibit marked sex differences in sensitivity to OGD, in part because of enhanced aromatization and estradiol formation in female cells.


Asunto(s)
Aromatasa/metabolismo , Astrocitos/fisiología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/enzimología , Western Blotting , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Hipoxia de la Célula/fisiología , Células Cultivadas , Femenino , Glucosa/deficiencia , Peróxido de Hidrógeno/toxicidad , Interleucina-1beta/toxicidad , Lipopolisacáridos/toxicidad , Masculino , Óxido Nítrico/toxicidad , Estrés Oxidativo/efectos de los fármacos , ARN/biosíntesis , ARN/aislamiento & purificación , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Caracteres Sexuales , Factor de Necrosis Tumoral alfa/toxicidad
19.
J Endocrinol ; 194(1): 153-60, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17592029

RESUMEN

The neuropeptides vasoactive intestinal peptide (VIP) and pituitary adenylate cyclase-activating polypeptide have long been considered as putative regulators of testicular functions. In vitro evidence suggests that VIP could play an important role in testosterone biosynthesis. However, the endogenous role of VIP on testicular functions remained to be demonstrated. In C57BL/6 mice exhibiting a complete disruption of the VIP gene, we first observed here that serum testosterone levels were lower than those of WT littermates. At the age of 4 months, this phenotype was accompanied with a reduction of expression of StAR and 3-beta-hydroxysteroid dehydrogenase (3beta-HSD) in the testis. In addition, serum levels of FSH but not LH were reduced in young knock-out (KO) males. Testicular anatomy also revealed a higher percentage of degenerated seminiferous tubules in the 4-month-old VIP KO animals when compared with WT. In 15-month-old animals, control males showed typical testicular aging, including a severe degeneration of seminiferous tubules, a dramatic decrease in serum levels of testosterone, and a reduction in StAR and 3beta-HSD gene expression. In age-matching VIP KO males, the levels of serum testosterone and steroidogenic enzymes were still very low. Interestingly, in contrast to that observed in young mice, testicular degeneration at 15 months was significantly less severe in VIP KO than WT mice. All together, these results suggest that 1) VIP is an important factor for regulating testosterone biosynthesis and FSH secretion and 2) VIP may influence testicular aging.


Asunto(s)
Envejecimiento/fisiología , Testículo/metabolismo , Testosterona/análisis , Péptido Intestinal Vasoactivo/genética , 3-Hidroxiesteroide Deshidrogenasas/genética , Animales , Secuencia de Bases , Hormona Folículo Estimulante/sangre , Expresión Génica , Hormona Luteinizante/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Fosfoproteínas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Testículo/citología , Testosterona/biosíntesis
20.
Brain Res ; 1110(1): 76-80, 2006 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-16872581

RESUMEN

The objective of this study was to examine the expression of receptors for androgen, estrogen, and progesterone in the fetal sheep brain during the critical period for sexual differentiation. We isolated mRNA from the hypothalamus-preoptic area (HPOA), amygdala (AMYG), medulla (MD), frontal cortex (FCTX) and olfactory bulbs (OB) of fetal sheep that were delivered on day 64 of gestation. Using a ribonuclease protection assay and species-specific cRNA probes, we measured mRNA expression levels of androgen receptor (AR), estrogen receptor alpha (ERalpha) and progesterone receptor (PR). ERalpha and AR mRNA were expressed in all of the tissues tested and highest in the HPOA. PR mRNA was measured in HPOA and AMYG only and was significantly higher in male than in female fetuses. We conclude that the fetal brain is a target site for circulating steroid hormones. These data have implications for the steroid dependent development of sexually dimorphic brain functions in sheep.


Asunto(s)
Encéfalo/metabolismo , Período Crítico Psicológico , Regulación del Desarrollo de la Expresión Génica/fisiología , Receptores de Esteroides/metabolismo , Diferenciación Sexual/fisiología , Animales , Encéfalo/anatomía & histología , Encéfalo/embriología , Embrión de Mamíferos , Femenino , Cabras , Masculino , ARN Mensajero/metabolismo , Receptores de Esteroides/clasificación , Receptores de Esteroides/genética , Ribonucleasas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA