Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Toxicol Sci ; 2024 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-38526210

RESUMEN

In avian embryos, xenoestrogens induce abnormalities in reproductive organs, particularly the testes and Müllerian ducts (MDs). However, the molecular mechanisms remain poorly understood. We investigated the effects of ethynylestradiol (EE2) exposure on gene expression associated with reproductive organ development in Japanese quail embryos. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) analysis revealed that the left testis containing ovary-like tissues following EE2 exposure highly expressed the genes for steroidogenic enzymes (P450scc, P45017α, lyase, and 3ß-HSD) and estrogen receptor-ß, compared to the right testis. No asymmetry was found in these gene expression without EE2. EE2 induced hypertrophy in female MDs and suppressed atrophy in male MDs on both sides. RNA sequencing analysis of female MDs showed 1,366 differentially expressed genes between developing left MD and atrophied right MD in the absence of EE2, and these genes were enriched in Gene Ontology terms related to organogenesis, including cell proliferation, migration and differentiation, and angiogenesis. However, EE2 reduced asymmetrically expressed genes to 21. RT-qPCR analysis indicated that genes promoting cell cycle progression and oncogenesis were more highly expressed in the left MD than in the right MD, but EE2 eliminated such asymmetric gene expression by increasing levels on the right side. EE2-exposed males showed overexpression of these genes in both MDs. This study reveals part of the molecular basis of xenoestrogen-induced abnormalities in avian reproductive organs, where EE2 may partly feminize gene expression in the left testis, developing as the ovotestis, and induce bilateral MD malformation by canceling asymmetric gene expression underlying MD development.

2.
J Appl Toxicol ; 44(5): 699-711, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38102769

RESUMEN

In ovo exposure to o,p'-dichloro-diphenyl-trichloroethane (o,p'-DDT) impairs reproduction by inducing malformation of the reproductive organs in birds, although the mechanism remains unclear. Here, we examined the effects of o,p'-DDT on the development of the reproductive organs, the expression of genes controlling sexual differentiation, and the plasma concentrations of testosterone and estradiol in Japanese quail embryos. o,p'-DDT-containing sesame oil was injected into the yolk sac on Embryonic Day (E) 3 at a dose of 500, 2,000, or 8,000 µg per egg. On E15, the reproductive organs were observed; the gonads and Müllerian ducts (MDs) were sampled to measure the mRNA of steroidogenic enzymes, sex steroid receptors, anti-Müllerian hormone (AMH), and AMH receptor 2 (AMHR2); blood samples were collected to assay plasma testosterone and estradiol levels; and the gonads were used for histological analysis. o,p'-DDT dose-dependently increased the prevalence of hypertrophic MDs in females and residual MDs in males. In female MDs, o,p'-DDT dose-dependently decreased estrogen receptor (ER) α, ERß, and AMHR2 mRNA expression. o,p'-DDT dose-dependently induced left-biased asymmetry of testis size, and ovary-like tissue was found in the left testis after exposure to 8,000 µg per egg o,p'-DDT, although asymmetric gene expression did not occur. o,p'-DDT did not affect ovarian tissue but did decrease 17α-hydroxylase/C17-20 lyase mRNA expression and dose-dependently increased ERß mRNA expression. o,p'-DDT decreased plasma testosterone concentrations in females. These findings suggest that o,p'-DDT induces hypertrophy of the MDs and ovarian tissue formation in the left testis. Abnormal MD development may be linked to altered gene expression for sensing estrogens and AMH signals.


Asunto(s)
Coturnix , Diferenciación Sexual , Animales , Masculino , Femenino , Coturnix/genética , Coturnix/metabolismo , Receptor beta de Estrógeno , DDT , Estradiol/metabolismo , Genitales , Testosterona , ARN Mensajero/genética
3.
J Neurosci ; 43(44): 7322-7336, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37722849

RESUMEN

The medial preoptic area (MPOA) is a sexually dimorphic region of the brain that regulates social behaviors. The sexually dimorphic nucleus (SDN) of the MPOA has been studied to understand sexual dimorphism, although the anatomy and physiology of the SDN is not fully understood. Here, we characterized SDN neurons that contribute to sexual dimorphism and investigated the mechanisms underlying the emergence of such neurons and their roles in social behaviors. A target-specific neuroanatomical study using transgenic mice expressing Cre recombinase under the control of Calb1, a gene expressed abundantly in the SDN, revealed that SDN neurons are divided into two subpopulations, GABA neurons projecting to the ventral tegmental area (VTA), where they link to the dopamine system (CalbVTA neurons), and GABA neurons that extend axons in the MPOA or project to neighboring regions (CalbnonVTA neurons). CalbVTA neurons were abundant in males, but were scarce or absent in females. There was no difference in the number of CalbnonVTA neurons between sexes. Additionally, we found that emergence of CalbVTA neurons requires two testicular androgen actions that occur first in the postnatal period and second in the peripubertal period. Chemogenetic analyses of CalbVTA neurons indicated a role in modulating sexual motivation in males. Knockdown of Calb1 in the MPOA reduced the intromission required for males to complete copulation. These findings provide strong evidence that a male-specific neural pathway from the MPOA to the VTA is organized by the two-step actions of testicular androgens for the modulation of sexually motivated behavior.SIGNIFICANCE STATEMENT The MPOA is a sexually dimorphic region of the brain that regulates social behaviors, although its sexual dimorphism is not fully understood. Here, we describe a population of MPOA neurons that contribute to the sexual dimorphism. These neurons only exist in masculinized brains, and they project their axons to the ventral tegmental area, where they link to the dopamine system. Emergence of such neurons requires two testicular androgen actions that occur first in the postnatal period and second in the peripubertal period. These MPOA neurons endow masculinized brains with a neural pathway from the MPOA to the ventral tegmental area and modulate sexually motivated behavior in males.


Asunto(s)
Andrógenos , Área Preóptica , Animales , Ratones , Femenino , Masculino , Área Preóptica/fisiología , Andrógenos/metabolismo , Área Tegmental Ventral , Dopamina/metabolismo , Vías Nerviosas , Ratones Transgénicos
4.
Gen Comp Endocrinol ; 314: 113917, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34555414

RESUMEN

Japanese quail (Coturnix japonica) is an avian model used to evaluate the reproductive and developmental toxicity of chemicals. The National Institute for Environmental Studies (NIES) of Japan established a strain of Japanese quail, NIES-L, which may be a better model because of its highly inbred characteristics. To understand sexual differentiation of the reproductive organs and the value of using NIES-L quails for avian toxicity assessment, we profiled estradiol and androgen plasma levels by enzyme-linked immunosorbent assay; the mRNA levels of estrogen receptor-α (ERα), ERß, and androgen receptor (AR) in the gonads, Müllerian ducts, Wolffian ducts; and the mRNA levels of steroidogenic enzymes, cholesterol side chain cleavage enzyme (P450scc), 17α-hydroxylase/C17-20 lyase (P45017α, lyase), 3ß-hydroxysteroid dehydrogenase (3ß-HSD), 17ß-hydroxysteroid dehydrogenase (17ß-HSD), and aromatase (P450arom), anti-Müllerian hormone (AMH), and AMH receptor type 2 (AMHR2) in the gonads of NIES-L Japanese quails on embryonic days 9, 12, and 15 using a real-time quantitative PCR method. The plasma estradiol concentration was higher in females than males on these embryonic days, but no sex difference was found in the plasma androgens. The mRNA levels of all examined steroidogenic enzymes were significantly higher in female than male embryos. In particular, the P450arom mRNA levels showed a striking sex difference: P450arom was expressed in female but not male gonads. In contrast, the AMH and AMHR2 mRNA levels in the gonads were higher in males than females. The ERα, ERß, and AR mRNA levels increased in the left female gonad and peaked on embryonic day 15, but not in the left and right male gonads; therefore, there was a female-biased sex difference. The ERα, ERß, and AR mRNA levels in the left Müllerian duct, but not in the right Müllerian duct, of females increased and peaked on embryonic day 15, which resulted in asymmetric mRNA levels. The Wolffian ducts expressed ERα, ERß, and AR in both sexes, and no sex difference or asymmetry of mRNA levels was found. The information obtained from this study helps elucidate the molecular endocrinological basis of sexual dimorphism formation of reproductive organs and clarify the value of NIES-L quails for toxicity assessment.


Asunto(s)
Coturnix , Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Regulación del Desarrollo de la Expresión Génica , Caracteres Sexuales , Diferenciación Sexual , Animales , Coturnix/genética , Coturnix/metabolismo , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/genética , Femenino , Genitales/metabolismo , Gónadas/metabolismo , Masculino , Diferenciación Sexual/genética
5.
Neurosci Lett ; 755: 135915, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-33905774

RESUMEN

The medial preoptic area, which plays an essential role in the control of sexual behavior in rats, contains a sexually dimorphic nucleus that consists of neurons expressing calbindin-D28 K (Calb) that is referred to as the CALB-SDN. The CALB-SDN is larger and contains more Calb neurons in males than in females. The physiological functions of the CALB-SDN are not fully understood; however, CALB-SDN neurons are activated during sexual behavior in males, suggesting that the male CALB-SDN is involved in regulation of sexual behavior. However, no information exists about the physiological functions of the female CALB-SDN. In the present study, we performed an immunohistochemical analysis of c-Fos, a neuronal activity marker, in the CALB-SDN of female and male rats that had copulated with conspecifics of the opposite sex to determine whether neurons of the female CALB-SDN are activated during copulation and whether the neuronal activity of the CALB-SDN differs between sexes. The numbers of c-Fos-immunoreactive cells with or without Calb-immunoreactivity (c-Fos+/Calb+ and c-Fos+/Calb- cells) were greater in the CALB-SDN of rats that had copulated than in rats that had not copulated in each sex. Although the number of Calb+ cells in the CALB-SDN was smaller in females than in males, the increase in the number of c-Fos+/Calb+ cells in the female CALB-SDN with copulation was comparable to that in the male CALB-SDN with copulation. The increase in the number of c-Fos+/Calb- cells in the CALB-SDN with copulation was more prominent in males than in females. These results suggest that CALB-SDN neurons are activated during copulation in both sexes. The patterns of neuronal activation in the CALB-SDN during copulation may differ between sexes.


Asunto(s)
Copulación/fisiología , Neuronas/metabolismo , Área Preóptica/metabolismo , Caracteres Sexuales , Animales , Calbindinas/metabolismo , Femenino , Masculino , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Wistar
6.
J Neuroendocrinol ; 33(3): e12961, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33675127

RESUMEN

Under severe calorie restriction (CR), the ghrelin-growth hormone axis in mice is involved in the maintenance of plasma glucose levels. Ghrelin, a stomach-derived acylated peptide, is up-regulated by the sympathetic nerve in the negative energy status. Central corticotrophin-releasing factor receptor (CRF-R) signalling stimulates the sympathetic tone. The present study aimed to examine the effect of central CRF-R signalling on the maintenance of plasma glucose concentrations in severe calorie-restricted mice with the involvement of ghrelin. Intracerebroventricular injections of urocorin-1 and urocorin-2, which are natural ligands for CRF-R1 and CRF-R2, elevated plasma ghrelin concentrations and ghrelin elevation with an i.c.v. injection of urocorin-1 was cancelled by atenolol (ß1 adrenergic receptor antagonist) administration. We then established a mice model of 60% CR and found that the administration of [d-Lys3]-GHRP-6 (a ghrelin receptor antagonist) in mice under 60% CR reduced the plasma glucose concentration more compared to the vehicle mice. Similarly, the atenolol injection in mice under 60% CR significantly reduced the plasma glucose concentration, which was rescued by the co-administration of ghrelin. An i.c.v. injection of the alpha helical CRH, a non-selective corticotrophin-releasing factor receptor antagonist, in mice under 60% CR significantly reduced the plasma glucose concentration, although the co-administration of α-helical CRH with ghrelin maintained plasma glucose levels. These results suggest that central CRF-R signalling is involved in the maintenance of plasma glucose levels in mice under severe CR via the sympathetic-ghrelin pathway.


Asunto(s)
Glucemia/metabolismo , Restricción Calórica , Ghrelina/fisiología , Receptores de Hormona Liberadora de Corticotropina/fisiología , Transducción de Señal/fisiología , Agonistas Adrenérgicos beta/farmacología , Animales , Atenolol/farmacología , Hormona Liberadora de Corticotropina/farmacología , Ghrelina/metabolismo , Inyecciones Intraventriculares , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Receptores de Ghrelina/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Sistema Nervioso Simpático/efectos de los fármacos
7.
Int J Mol Sci ; 22(2)2021 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-33430368

RESUMEN

Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by impaired social communication, poor social interactions, and repetitive behaviors. We aimed to examine autism-like behaviors and related gene expressions in rats exposed to diesel exhaust (DE)-origin secondary organic aerosol (DE-SOA) perinatally. Sprague-Dawley pregnant rats were exposed to clean air (control), DE, and DE-SOA in the exposure chamber from gestational day 14 to postnatal day 21. Behavioral phenotypes of ASD were investigated in 10~13-week-old offspring using a three-chambered social behavior test, social dominance tube test, and marble burying test. Prefrontal cortex was collected to examine molecular analyses including neurological and immunological markers and glutamate concentration, using RT-PCR and ELISA methods. DE-SOA-exposed male and female rats showed poor sociability and social novelty preference, socially dominant behavior, and increased repetitive behavior. Serotonin receptor (5-HT(5B)) and brain-derived neurotrophic factor (BDNF) mRNAs were downregulated whereas interleukin 1 ß (IL-ß) and heme oxygenase 1 (HO-1) mRNAs were upregulated in the prefrontal cortex of male and female rats exposed to DE-SOA. Glutamate concentration was also increased significantly in DE-SOA-exposed male and female rats. Our results indicate that perinatal exposure to DE-SOA may induce autism-like behavior by modulating molecules such as neurological and immunological markers in rats.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Trastorno del Espectro Autista/fisiopatología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Emisiones de Vehículos/toxicidad , Aerosoles/toxicidad , Animales , Trastorno del Espectro Autista/inducido químicamente , Factor Neurotrófico Derivado del Encéfalo/genética , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Hemo-Oxigenasa 1/genética , Humanos , Interleucina-1beta/genética , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/genética , Ratas , Ratas Sprague-Dawley , Receptores de Serotonina/genética
8.
Front Neurosci ; 14: 797, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32848568

RESUMEN

Testicular androgens during the perinatal period play an important role in the sexual differentiation of the brain of rodents. Testicular androgens transported into the brain act via androgen receptors or are the substrate of aromatase, which synthesizes neuroestrogens that act via estrogen receptors. The latter that occurs in the perinatal period significantly contributes to the sexual differentiation of the brain. The preoptic area (POA) and the bed nucleus of the stria terminalis (BNST) are sexually dimorphic brain regions that are involved in the regulation of sex-specific social behaviors and the reproductive neuroendocrine system. Here, we discuss how neuroestrogens of testicular origin act in the perinatal period to organize the sexually dimorphic structures of the POA and BNST. Accumulating data from rodent studies suggest that neuroestrogens induce the sex differences in glial and immune cells, which play an important role in the sexually dimorphic formation of the dendritic synapse patterning in the POA, and induce the sex differences in the cell number of specific neuronal cell groups in the POA and BNST, which may be established by controlling the number of cells dying by apoptosis or the phenotypic organization of living cells. Testicular androgens in the peripubertal period also contribute to the sexual differentiation of the POA and BNST, and thus their aromatization to estrogens may be unnecessary. Additionally, we discuss the notion that testicular androgens that do not aromatize to estrogens can also induce significant effects on the sexually dimorphic formation of the POA and BNST.

9.
Psychoneuroendocrinology ; 120: 104792, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32653768

RESUMEN

Maternally experienced female rats show high maternal behavior performance for a long time after acquisition of maternal experience, although the mechanisms responsible for the retention of maternal behavior are not well understood. The medial preoptic area (MPOA) plays an important role in the onset and maintenance of maternal behavior in female rats. We aimed to determine whether maternal experience affects the glutamatergic system in the MPOA for the retention of maternal behavior in female rats. First, to determine the effects of maternal experience in the postpartum period on dendritic spines, which are the postsynaptic component of excitatory glutamatergic neurotransmission, we examined the number of dendritic spines on MPOA neurons of primiparous mothers that had experienced mothering until weaning (sufficiently experienced mothers) and of primiparous mothers that were separated from their pups on the day of parturition (insufficiently experienced mothers). The number of mushroom spines, but not other types of spine, was significantly greater in the sufficiently experienced mothers compared with that in the insufficiently experienced mothers. Next, to determine the effects of maternal experience in the postpartum period on the expression of ionotropic glutamate receptors, we measured the mRNA levels of AMPA receptor subunits (GluA1-A4) and NMDA receptor subunits (GluN1, GluN2A-2D) in the MPOA of primiparous female rats that were kept with pups until brain sampling. As a result, we found that the mRNA levels of GluA3 and GluN2B were significantly higher in primiparous females on the day of weaning compared with those in primiparous females on the day of parturition. Additionally, we examined the effects of CNQX, an AMPA receptor antagonist, and MK-801, an NMDA receptor antagonist, injected into the MPOA on maternal behavior in maternally experienced primiparous female rats. Maternal behavioral activity was significantly reduced when CNQX or MK-801 was injected into the MPOA. These findings indicate that long-term maternal experience in the postpartum period up-regulates glutamatergic neurotransmission by increasing the number of mushroom spines and glutamate receptor expression, which may be involved in the retention of maternal behavior in maternally experienced female rats.


Asunto(s)
Conducta Materna/fisiología , Área Preóptica/fisiología , Receptores Ionotrópicos de Glutamato/metabolismo , Animales , Espinas Dendríticas/metabolismo , Aminoácidos Excitadores/metabolismo , Femenino , Ácido Glutámico/metabolismo , Madres , Neuronas/metabolismo , Periodo Posparto/metabolismo , Periodo Posparto/fisiología , Área Preóptica/metabolismo , Ratas , Ratas Wistar , Receptores AMPA/metabolismo , Receptores Ionotrópicos de Glutamato/análisis , Receptores Ionotrópicos de Glutamato/genética , Receptores de N-Metil-D-Aspartato/metabolismo
10.
Neuroscience ; 438: 182-197, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32387645

RESUMEN

Two types of nuclear estrogen receptors, ERα and ERß, have been shown to be differentially involved in the regulation of various types of behaviors. Due to a lack of tools for identifying ERß expression, detailed anatomical distribution and neurochemical characteristics of ERß expressing cells and cellular co-expression with ERα remain unclear. We have generated transgenic mice ERß-RFPtg, in which RFP was inserted downstream of ERß BAC promotor. We verified RFP signals as ERß by confirming: (1) high ERß mRNA levels in RFP-expressing cells collected by fluorescence-activated cell sorting; and (2) co-localization of ERß mRNA and RFP proteins in the paraventricular nucleus (PVN). Strong ERß-RFP signals were found in the PVN, medial preoptic area (MPOA), bed nucleus of the stria terminalis, medial amygdala (MeA), and dorsal raphe nucleus (DRN). In the MPOA and MeA, three types of cell populations were identified; those expressing both ERα and ERß, and those expressing exclusively either ERα or ERß. The majority of PVN and DRN cells expressed only ERß-RFP. Further, ERß-RFP positive cells co-expressed oxytocin in the PVN, and tryptophan hydroxylase 2 and progesterone receptors in the DRN. In the MeA, some ERß-RFP positive cells co-expressed oxytocin receptors. These findings collectively suggest that ERß-RFPtg mice can be a powerful tool for future studies on ERß function in the estrogenic regulation of social behaviors.


Asunto(s)
Receptor alfa de Estrógeno , Receptor beta de Estrógeno , Animales , Encéfalo/metabolismo , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno/genética , Ratones , Ratones Transgénicos , Núcleo Hipotalámico Paraventricular/metabolismo , Receptores de Estrógenos/metabolismo
11.
Endocrinology ; 161(6)2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32303738

RESUMEN

The calbindin-sexually dimorphic nucleus (CALB-SDN) and calbindin-principal nucleus of the bed nucleus of the stria terminalis (CALB-BNSTp) show male-biased sex differences in calbindin neuron number. The ventral part of the BNSTp (BNSTpv) exhibits female-biased sex differences in noncalbindin neuron number. We previously reported that prepubertal gonadectomy disrupts the masculinization of the CALB-SDN and CALB-BNSTp and the feminization of the BNSTpv. This study aimed to determine the action mechanisms of testicular androgens on the masculinization of the CALB-SDN and CALB-BNSTp and whether ovarian estrogens are the hormones that have significant actions in the feminization of the BNSTpv. We performed immunohistochemical analyses of calbindin and NeuN, a neuron marker, in male mice orchidectomized on postnatal day 20 (PD20) and treated with cholesterol, testosterone, estradiol, or dihydrotestosterone during PD20-70, female mice ovariectomized on PD20 and treated with cholesterol or estradiol during PD20-70, and PD70 mice gonadectomized on PD56. Calbindin neurons number in the CALB-SDN and CALB-BNSTp in males treated with testosterone or dihydrotestosterone, but not estradiol, was significantly larger than that in cholesterol-treated males. Noncalbindin neuron number in the BNSTpv in estradiol-treated females was significantly larger than that in cholesterol-treated females. Gonadectomy on PD56 had no significant effect on neuron numbers. Additionally, an immunohistochemical analysis revealed the expression of androgen receptors in the CALB-SDN and CALB-BNSTp of PD30 males and estrogen receptors-α in the BNSTpv of PD30 females. These results suggest that peripubertal testicular androgens act to masculinize the CALB-SDN and CALB-BNSTp without aromatization, and peripubertal ovarian estrogens act to feminize the BNSTpv.


Asunto(s)
Encéfalo/metabolismo , Hormonas Esteroides Gonadales/metabolismo , Ratones/metabolismo , Pubertad/metabolismo , Caracteres Sexuales , Animales , Encéfalo/crecimiento & desarrollo , Calbindinas/metabolismo , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Estrógenos/metabolismo , Femenino , Masculino , Ratones/genética , Ratones/crecimiento & desarrollo , Neuronas/metabolismo , Ovario/crecimiento & desarrollo , Ovario/metabolismo , Testículo/metabolismo , Testosterona/metabolismo
12.
Environ Int ; 138: 105662, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32203809

RESUMEN

Developmental exposure to environmental chemicals with estrogen-like activity is suspected to permanently impair women's health. In this study, a mouse model was used to evaluate whether tris(2,6-dimethylphenyl) phosphate (TDMPP), a chemical with a putative estrogen-like action, impairs sexual differentiation of the brain. Either TDMPP and 17ß-estradiol (E2) as positive controls or sesame oil as a negative control were administered subcutaneously to dams from gestational day (GD) 14 to parturition, and to pups from postnatal day (PND) 0 to 9. Precocious puberty, irregular estrous cycles, and a lowered lordosis response were found in the TDMPP- and E2-treated groups. A certain amount of TDMPP and its metabolites in the perinatal brain and the masculinization of sexual dimorphic nuclei in the hypothalamus of female mice after treatment were also detected. The experimental evidence demonstrates that TDMPP directly enters the fetal and neonatal brain, thereby inducing changes of sex-related brain structures and impairing female reproductive functions.


Asunto(s)
Estradiol , Fosfatos , Animales , Estrona , Femenino , Desarrollo Fetal , Ratones , Embarazo
13.
Neurosci Biobehav Rev ; 110: 46-59, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-30392880

RESUMEN

It has long been known that the estrogen, 17ß-estradiol (17ß-E), plays a central role for female reproductive physiology and behavior. Numerous studies have established the neurochemical and molecular basis of estrogenic induction of female sexual behavior, i.e., lordosis, in animal models. In addition, 17ß-E also regulates male-type sexual and aggressive behavior. In males, testosterone secreted from the testes is irreversibly aromatized to 17ß-E in the brain. We discuss the contribution of two nuclear receptor isoforms, estrogen receptor (ER)α and ERß to the estrogenic regulation of sexually dimorphic brain formation and sex-typical expression of these social behaviors. Furthermore, 17ß-E is a key player for social behaviors such as social investigation, preference, recognition and memory as well as anxiety-related behaviors in social contexts. Recent studies also demonstrated that not only nuclear receptor-mediated genomic signaling but also membrane receptor-mediated non-genomic actions of 17ß-E may underlie the regulation of these behaviors. Finally, we will discuss how rapidly developing research tools and ideas allow us to investigate estrogenic action by emphasizing behavioral neural networks.


Asunto(s)
Estrógenos/metabolismo , Memoria/fisiología , Reconocimiento en Psicología/fisiología , Conducta Social , Animales , Estrógenos/farmacología , Humanos , Memoria/efectos de los fármacos , Conducta Sexual/efectos de los fármacos , Conducta Sexual/fisiología , Conducta Sexual Animal/fisiología
14.
Psychoneuroendocrinology ; 108: 1-13, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31174081

RESUMEN

The medial preoptic nucleus (MPN) plays an essential role in the control of male sexual behavior. In rats, the central part of the MPN (MPNc) contains a sexually dimorphic nucleus exhibiting male-biased morphological sex differences. Although it has been suggested that the MPNc of male rats functions to induce sexual arousal, the mechanisms by which male rats are sexually aroused to successfully achieve copulation are poorly understood. We recently showed that increased neuronal activity in the MPNc of male rats during copulation is higher at their first copulation compared with later copulations, indicating that a plastic change in excitatory synaptic transmission occurs with copulatory experience. In this study, we tested the hypothesis that changes to dendritic spines at structural and molecular levels occur following copulatory experience. First, we examined the effects of at least two copulations on the morphology of dendrites and spines in the MPNc and in the lateral and medial parts of the MPN (MPNlm) of male rats. In the MPNc, the total number of dendrites and their branches, and the surface area of dendrites were not significantly affected by copulation. However, the copulatory experience, specifically experience of ejaculation, significantly reduced the density of mushroom spines but not of filopodia, thin or stubby spines in the MPNc. In the MPNlm, the copulatory experience, specifically experience of ejaculation, significantly increased the surface area of dendrites, although there was no significant effect of copulation on spine density. Next, we measured the mRNA levels of genes encoding actin-binding proteins related to spinogenesis after male rats had copulated for their first and second times. Copulatory stimuli, especially stimuli from ejaculation, significantly reduced the mRNA levels of drebrin A and spinophilin in the MPNc but not in the MPNlm. These results indicate that copulatory experiences, especially experience of ejaculation, reduce spine density in the MPNc of male rats, which may result, in part, from downregulation of genes encoding actin-binding proteins.


Asunto(s)
Copulación/fisiología , Espinas Dendríticas/metabolismo , Área Preóptica/metabolismo , Animales , Dendritas/genética , Dendritas/metabolismo , Espinas Dendríticas/genética , Eyaculación , Expresión Génica/genética , Masculino , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Plasticidad Neuronal/genética , Neuronas/metabolismo , Neuropéptidos/genética , Neuropéptidos/metabolismo , Área Preóptica/fisiología , Ratas , Ratas Wistar , Conducta Sexual Animal/fisiología
15.
Biol Sex Differ ; 10(1): 10, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30782210

RESUMEN

Following publication of the original article [1], we noticed a number of errors.

16.
Biol Sex Differ ; 10(1): 6, 2019 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-30691514

RESUMEN

BACKGROUND: The bed nucleus of the stria terminalis (BNST) contains the highest density of corticotropin-releasing factor (CRF)-producing neurons in the brain. CRF-immunoreactive neurons show a female-biased sexual dimorphism in the dorsolateral BNST in the rat. Since CRF neurons cannot be immunostained clearly with available CRF antibodies in the mouse, we used a mouse line, in which modified yellow fluorescent protein (Venus) was inserted to the CRF gene, and the Neo cassette was removed, to examine the morphological characteristics of CRF neurons in the dorsolateral BNST. Developmental changes of CRF neurons were examined from postnatal stages to adulthood. Gonadectomy (GDX) was carried out in adult male and female mice to examine the effects of sex steroids on the number of CRF neurons in the dorsolateral BNST. METHODS: The number of Venus-expressing neurons, stained by immunofluorescence, was compared between male and female mice over the course of development. GDX was carried out in adult mice. Immunohistochemistry, in combination with Nissl staining, was carried out, and the effects of sex or gonadal steroids were examined by estimating the number of Venus-expressing neurons, as well as the total number of neurons or glial cells, in each BNST subnucleus, using a stereological method. RESULTS: Most Venus-expressing neurons co-expressed Crf mRNA in the dorsolateral BNST. They constitute a group of neurons without calbindin immunoreactivity, which makes a contrast to the principal nucleus of the BNST that is characterized by calbindin immunostaining. In the dorsolateral BNST, the number of Venus-expressing neurons increased across developmental stages until adulthood. Sexual difference in the number of Venus-expressing neurons was not evident by postnatal day 5. In adulthood, however, there was a significant female predominance in the number of Venus expressing neurons in two subnuclei of the dorsolateral BNST, i.e., the oval nucleus of the BNST (ovBNST) and the anterolateral BNST (alBNST). The number of Venus-expressing neurons was smaller significantly in ovariectomized females compared with proestrous females in either ovBNST or alBNST, and greater significantly in orchiectomized males compared with gonadally intact males in ovBNST. The total number of neurons was also greater significantly in females than in males in ovBNST and alBNST, but it was not affected by GDX. CONCLUSION: Venus-expressing CRF neurons showed female-biased sexual dimorphism in ovBNST and alBNST of the mouse. Expression of Venus in these subnuclei was controlled by gonadal steroids.


Asunto(s)
Hormona Liberadora de Corticotropina/metabolismo , Neuronas/metabolismo , Núcleos Septales/metabolismo , Caracteres Sexuales , Animales , Castración , Hormona Liberadora de Corticotropina/genética , Femenino , Masculino , Ratones Transgénicos , Neuroglía/metabolismo , ARN Mensajero/metabolismo
17.
Neurosci Lett ; 694: 80-85, 2019 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-30472357

RESUMEN

Male rats rarely show lordosis, a female sexual behavior, because of strong inhibition of the behavior in the lateral septum. Because neonatal treatment with estradiol (E2) in female rats decreases lordosis, it is believed that the lateral septum is a target of E2 action to defeminize or masculinize the lordosis-inhibiting system. Here, we tested the hypothesis that disruption of the lateral septum before E2 treatment prevents the effect of neonatal E2 on lordosis. Female rats that underwent radiofrequency-induced septal lesions or sham operation on postnatal day 4 (PD4, day of birth = PD1) were subcutaneously injected with E2 or sesame oil vehicle alone on PD5. Vaginal opening and smears were checked. After sexual maturation, lordosis tests were performed. The effects of neonatal septal lesions on lordosis in male rats were also observed. Sham-operated and E2-treated female rats showed a reduction in lordosis and irregular estrous cycles. Conversely, septal lesioned and E2-treated females exhibited higher levels of lordosis, although their estrous cycles were irregular. These results suggest that neonatal septal lesions prevent females from being behaviorally defeminized by neonatal E2. Additionally, neonatally septal lesioned males displayed higher levels of lordosis than sham-operated males. These results suggest that E2, which is produced by the aromatization of testicular testosterone in the neonatal period, acts on the lateral septum to organize the lordosis-inhibiting system.


Asunto(s)
Estradiol/fisiología , Postura , Núcleos Septales/fisiología , Caracteres Sexuales , Conducta Sexual Animal/fisiología , Animales , Animales Recién Nacidos , Estradiol/administración & dosificación , Ciclo Estral , Femenino , Masculino , Ratas Wistar , Núcleos Septales/efectos de los fármacos
18.
Endocrinology ; 159(12): 3993-4005, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30371765

RESUMEN

The central part of the medial preoptic nucleus (MPNc) is associated with sexual arousal induction in male rats. However, it is largely unclear how males are sexually aroused and achieve their first copulation. We previously reported that more MPNc neurons activate during the first copulation than the second copulation. In this study, to explore the molecules responsible for sexual arousal induction, we performed DNA microarray of the MPNc in sexually naive males and males after they copulated for their first and second times. We then performed quantitative PCR analyses to validate the results of the DNA microarray. Six genes were identified. Their expression increased following copulation and was higher in males after they copulated for the first time than after the second time. The genes encode transcription factors (Fos, Nfil3, and Nr4a3), a serine/threonine kinase (Sik1), an antioxidant protein (Srxn1), and a neuropeptide precursor VGF (Vgf), which may be the candidate genes responsible for sexual arousal induction. We examined the effects of Vgf knockdown in the MPNc on sexual partner preference and sexual behavior in sexually inexperienced and experienced males to determine the role of VGF in sexual arousal induction. A preference for estrous female rats was reinforced, and the latency of mount and intromission became short after sexually inexperienced males copulated for the first time. However, Vgf knockdown disrupted these phenomena. Vgf knockdown did not have any significant effect in sexually experienced males. VGF-derived neuropeptides presumably serve as an effector molecule to increase sexual activity following sexual arousal induction.


Asunto(s)
Nivel de Alerta/genética , Neuropéptidos/fisiología , Área Preóptica/metabolismo , Conducta Sexual Animal/fisiología , Animales , Copulación/fisiología , Femenino , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Masculino , Neuropéptidos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas , Ratas Transgénicas , Ratas Wistar , Factores Sexuales
19.
Neurosci Lett ; 685: 155-159, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30170041

RESUMEN

The medial preoptic area (MPN) plays an important role in the control of male sexual behavior. In rats, the central part of the MPN (MPNc) is sexually dimorphic and contains a sexually dimorphic nucleus composed of neurons expressing calbindin-D28 K (CALB-SDN). Although the functions of the MPNc are not well understood, surgical destruction of the MPNc adversely affects the performance of sexual behavior in sexually naive males, but not in sexually experienced males, supporting the notion that the MPNc changes functionally with sexual experience. In this study, we aimed to determine the effects of sexual experience on the neuronal activity of the MPNc and CALB-SDN. Sexual behavior in sexually inexperienced males that had no experience of ejaculation, and experienced males that had ejaculated once was observed. After they displayed sexual behavior, the brains were sampled, and immunohistochemical analysis of c-Fos, a neuronal activity marker, in the MPNc and CALB-SDN was performed. The numbers of c-Fos-immunopositive cells with or without calbindin-D28K-immunoreactivity increased significantly in the MPNc and CALB-SDN following ejaculation in both sexually inexperienced and experienced males, although the numbers did not change significantly with exposure to estrous female odors, the first mount, and the first intromission before and after the first ejaculation. We further found that the number of c-Fos-immunopositive and calbindin-D28K-immunonegative cells in the MPNc, but not in the CALB-SDN, was significantly smaller in sexually experienced males than in sexually inexperienced males. These results suggest that a population of MPNc neurons, which is located outside the CALB-SDN and do not express calbindin-D28 K, are activated during the first copulation and then silent after acquisition of sexual experience.


Asunto(s)
Neuronas/metabolismo , Área Preóptica/metabolismo , Conducta Sexual Animal/fisiología , Conducta Sexual/fisiología , Animales , Eyaculación/fisiología , Masculino , Proteínas Proto-Oncogénicas c-fos/metabolismo , Caracteres Sexuales
20.
Artículo en Inglés | MEDLINE | ID: mdl-29770127

RESUMEN

Sex steroids play a major role in sexually dimorphic brain development during not only the perinatal period but also the pubertal period. We previously showed that, in male mice, the estrogen receptor-α (Esr1) and aromatase (Cyp19a1) genes are essential to the sexually dimorphic formation of the anteroventral periventricular nucleus (AVPV) and the principal nucleus of the bed nucleus of the stria terminalis (BNSTp), but the estrogen receptor-ß (Esr2) gene is not necessary. We also showed that the androgen receptor (Ar) gene is essential to the sexually dimorphic formation of the BNSTp. These genes are expressed in the AVPV and BNSTp of perinatal mice. However, it remains unknown whether these genes are expressed in the AVPV and BNSTp during puberty, and whether the expression, if any, differs by sex, age, and brain region. Here, we dissected the AVPV and BNSTp from Nissl-stained brain sections of male and female mice on postnatal day (PD) 20 (prepuberty), PD30 (puberty onset in females), PD40 (puberty onset in males), and PD60 (young adult) using a laser microdissection system. We then examined the mRNA levels of Esr1, Esr2, Cyp19a1, and Ar in these brain regions. In the AVPV, Esr1 mRNA levels were greater in females than males during PD20-60. Esr2 and Ar mRNA expressions did not differ between sexes. Ar mRNA levels were higher at PD30 than PD20. Cyp19a1 mRNA was not detected in the AVPV at PD20-60. In the BNSTp, Esr1 and Esr2 mRNA levels were higher in females than in males during PD20-60, although the mRNA levels of Cyp19a1 and Ar did not differ between sexes. Additionally, we revealed that orchiectomy at PD20 induced a failure of normal formation of the male BNSTp and testosterone replacement in the prepubertal period rescued the effect of orchiectomy at PD20. Taken together, it is suggested that pubertal testosterone transported to the AVPV is not converted to estradiol there and does not act via ESR1 and ESR2. By contrast, the formation of the male BNSTp may be affected by testicular testosterone during puberty via AR and/or via ESR1 after conversion to estradiol by CYP19A1.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...