Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Neuroendocrinol ; 35(5): e13284, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37157154

RESUMO

In goats, early exposure of spring-born females to sexually active bucks induces an early puberty onset assessed by the first ovulation. This effect is found when females are continuously exposed well before the male breeding season starting in September. The first aim of this study was to evaluate whether a shortened exposure of females to males could also lead to early puberty. We assessed the onset of puberty in Alpine does isolated from bucks (ISOL), exposed to wethers (CAS), exposed to intact bucks from the end of June (INT1), or mid-August (INT2). Intact bucks became sexually active in mid-September. At the beginning of October, 100% of INT1 and 90% of INT2 exposed does ovulated, in contrast to the ISOL (0%) and CAS (20%) groups. This demonstrated that contact with males that become sexually active is the main factor prompting precocious puberty in females. Furthermore, a reduced male exposure during a short window before the breeding season is sufficient to induce this phenomenon. The second aim was to investigate the neuroendocrine changes induced by male exposure. We found a significant increase in kisspeptin immunoreactivity (fiber density and number of cell bodies) in the caudal part of the arcuate nucleus of INT1 and INT2 exposed females. Thus, our results suggest that sensory stimuli from sexually active bucks (e.g., chemosignals) may trigger an early maturation of the ARC kisspeptin neuronal network leading to gonadotropin-releasing hormone secretion and first ovulation.


Assuntos
Núcleo Arqueado do Hipotálamo , Kisspeptinas , Masculino , Feminino , Animais , Maturidade Sexual , Hormônio Liberador de Gonadotropina , Cabras , Neurônios
2.
Reprod Fertil Dev ; 35(4): 307-320, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36593258

RESUMO

CONTEXT: Mammalian target of rapamycin complex 1 (mTORC1) is an essential sensor that regulates fundamental biological processes like cell growth, proliferation and energy metabolism. The treatment of disease by sirolimus, a mTORC1 inhibitor, causes adverse effects, such as female fertility disorders. AIMS: The objective of the study was to decipher the reproductive consequences of a downregulation of mTORC1 in the hypothalamus. METHODS: The reduced expression of mTORC1 was induced after intracerebroventricular injection of lentivirus expressing a short hairpin RNA (shRNA) against regulatory associated protein of TOR (raptor) in adult female mice (ShRaptor mice). KEY RESULTS: The ShRaptor mice were fertile and exhibited a 15% increase in the litter size compared with control mice. The histological analysis showed an increase in antral, preovulatory follicles and ovarian cysts. In the hypothalamus, the GnRH mRNA and FSH levels in ShRaptor mice were significantly elevated. CONCLUSIONS: These results support the hypothesis that mTORC1 in the central nervous system participates in the regulation of female fertility and ovarian function by influencing the GnRH neuronal activity. IMPLICATIONS: These results suggest that a lower mTORC1 activity directly the central nervous system leads to a deregulation in the oestrous cycle and an induction of ovarian cyst development.


Assuntos
Cistos Ovarianos , Aves Predatórias , Feminino , Animais , Camundongos , Humanos , Serina-Treonina Quinases TOR/metabolismo , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Fatores de Transcrição/metabolismo , RNA Interferente Pequeno , Hipotálamo/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Aves Predatórias/genética , Aves Predatórias/metabolismo , Mamíferos/genética
3.
Artigo em Inglês | MEDLINE | ID: mdl-33013709

RESUMO

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


Assuntos
Disruptores Endócrinos/administração & dosagem , Inseticidas/administração & dosagem , Metoxicloro/administração & dosagem , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Reprodução/efeitos dos fármacos , Comportamento Sexual Animal/efeitos dos fármacos , Animais , Ciclo Estral/efeitos dos fármacos , Feminino , Kisspeptinas/metabolismo , Camundongos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Área Pré-Óptica/efeitos dos fármacos , Área Pré-Óptica/metabolismo , Maturidade Sexual/efeitos dos fármacos
4.
Psychoneuroendocrinology ; 109: 104387, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31465941

RESUMO

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.


Assuntos
Encéfalo/efeitos dos fármacos , Hormônios Gonadais/metabolismo , Caracteres Sexuais , Tonsila do Cerebelo/metabolismo , Animais , Encéfalo/diagnóstico por imagem , Castração/métodos , Estradiol/farmacologia , Feminino , Hormônios Gonadais/fisiologia , Substância Cinzenta/diagnóstico por imagem , Substância Cinzenta/efeitos dos fármacos , Hipotálamo/metabolismo , Imageamento por Ressonância Magnética/métodos , Masculino , Ovinos , Testosterona/farmacologia
5.
Chemosphere ; 221: 573-582, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30660913

RESUMO

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


Assuntos
Dietilexilftalato/toxicidade , Poluentes Ambientais/toxicidade , Sistemas Neurossecretores/efeitos dos fármacos , Fenóis/toxicidade , Comportamento Sexual Animal/efeitos dos fármacos , Animais , Feminino , Kisspeptinas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Testosterona/metabolismo
6.
J Neuroendocrinol ; 31(2): e12681, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30585662

RESUMO

Both systemic and local production contribute to the concentration of steroids measured in the brain. This idea was originally based on rodent studies and was later extended to other species, including humans and birds. In quail, a widely used model in behavioural neuroendocrinology, it was demonstrated that all enzymes needed to produce sex steroids from cholesterol are expressed and active in the brain, although the actual concentrations of steroids produced were never investigated. We carried out a steroid profiling in multiple brain regions and serum of sexually mature male and female quail by gas chromatography coupled with mass spectrometry. The concentrations of some steroids (eg, corticosterone, progesterone and testosterone) were in equilibrium between the brain and periphery, whereas other steroids (eg, pregnenolone (PREG), 5α/ß-dihydroprogesterone and oestrogens) were more concentrated in the brain. In the brain regions investigated, PREG sulphate, progesterone and oestrogen concentrations were higher in the hypothalamus-preoptic area. Progesterone and its metabolites were more concentrated in the female than the male brain, whereas testosterone, its metabolites and dehydroepiandrosterone were more concentrated in males, suggesting that sex steroids present in quail brain mainly depend on their specific steroidogenic pathways in the ovaries and testes. However, the results of castration experiments suggested that sex steroids could also be produced in the brain independently of the peripheral source. Treatment with testosterone or oestradiol restored the concentrations of most androgens or oestrogens, respectively, although penetration of oestradiol in the brain appeared to be more limited. These studies illustrate the complex interaction between local brain synthesis and the supply from the periphery for the steroids present in the brain that are either directly active or represent the substrate of centrally located enzymes.


Assuntos
Encéfalo/metabolismo , Codorniz/fisiologia , Caracteres Sexuais , Esteroides/sangue , Esteroides/metabolismo , 20-alfa-Di-Hidroprogesterona/sangue , 20-alfa-Di-Hidroprogesterona/metabolismo , 5-alfa-Di-Hidroprogesterona/sangue , 5-alfa-Di-Hidroprogesterona/metabolismo , Animais , Castração , Corticosterona/sangue , Corticosterona/metabolismo , Estrogênios/sangue , Estrogênios/metabolismo , Feminino , Hipotálamo/metabolismo , Masculino , Pregnenolona/sangue , Pregnenolona/metabolismo , Área Pré-Óptica/metabolismo , Testosterona/sangue , Testosterona/metabolismo
7.
Horm Behav ; 106: 81-92, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30308180

RESUMO

In rodents, early exposure to adult male is well known to induce an early puberty in females (Vandenbergh effect). This phenomenon has been less studied in other mammals. In goats, despite our extensive knowledge about the "male-effect" phenomenon in adults (i.e. ovulation induced by the introduction of the male during the anestrous), there are few data on the consequences of an early exposure of females to males. Here, we evaluated the puberty onset of young alpine goats when raised since weaning with intact bucks (INT), with castrated bucks (CAS) or isolated from bucks (ISOL). The INT group had the first ovulation 1.5 month before the two other groups. Despite the earlier puberty the INT group of females had normal and regular ovarian cycles. Morphological study of the genital tract showed that at 6 months, uterus of INT goats was 40% heavier than CAS and ISOL goats. Moreover, INT females had a myometrium significantly thicker and INT was the only group having corpora lutea. In our study, INT females were pubescent in the month following the entry of bucks into the breeding season, suggesting that only sexually active bucks provide the signal responsible for puberty acceleration. By removing direct contact with the bucks, we showed that somatosensory interactions were dispensable for an early puberty induction. Finally, no difference in the GnRH network (fiber density and number of synaptic appositions) can be detected between pubescent and non-pubescent females, suggesting that the male stimulations triggering puberty onset act probably on upstream neuronal networks, potentially on kisspeptin neurons.


Assuntos
Sinais (Psicologia) , Cabras/fisiologia , Ovulação/fisiologia , Comportamento Sexual Animal/fisiologia , Maturidade Sexual/fisiologia , Comportamento Social , Anestro/fisiologia , Animais , Feminino , Hormônio Liberador de Gonadotropina/sangue , Masculino , Estações do Ano
8.
Sci Rep ; 8(1): 2988, 2018 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-29445187

RESUMO

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


Assuntos
Corte , Poluentes Ambientais/metabolismo , Neurônios/fisiologia , Fenóis/metabolismo , Vocalização Animal/fisiologia , Animais , Ansiedade , Disruptores Endócrinos , Estradiol/metabolismo , Locomoção , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Condução Nervosa , Condutos Olfatórios , Receptores Androgênicos/metabolismo , Receptores de Estrogênio/metabolismo , Testosterona/metabolismo
9.
Physiol Behav ; 165: 173-8, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27475456

RESUMO

In small ungulates such as sheep or goats, the introduction of a male among a group of anovulatory females during the anestrus season leads to the reactivation of the gonadotrope axis and ovulation, a phenomenon known as the 'male effect'. In goats, our previous studies have demonstrated the importance of male sexual activity for an efficient reactivation of the gonadotrope axis assessed through ovulation and blood LH pulsatility. In the present experiment, we assessed whether the level of male sexual activity would also induce differential activation of two brain regions of key importance for the reactivation of GnRH activity, namely the medial preoptic area and the hypothalamic arcuate nucleus. In both structures, we observed a differential activation of Fos in females, depending on the level of buck sexual activity. Indeed, goats unexposed to males showed low levels of expression of Fos while those exposed to sexually inactive bucks showed an intermediate level of Fos expression. Finally, the highest level of Fos expression was found in females exposed to sexually active males. However, and contrary to our initial hypothesis, we were not able to find any specific activation of kisspeptin cells in the arcuate nucleus following the introduction of highly sexually active males. As a whole, these results demonstrate that the level of male sexual activity is a key factor to stimulate brain regions involved in the control of the gonadotrope axis in the context of the male effect in goats.


Assuntos
Anestro/fisiologia , Núcleo Arqueado do Hipotálamo/fisiologia , Área Pré-Óptica/fisiologia , Caracteres Sexuais , Comportamento Sexual/fisiologia , Análise de Variância , Animais , Feminino , Cabras , Hormônio Liberador de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Masculino , Proteínas Oncogênicas v-fos/metabolismo
10.
Horm Behav ; 80: 1-9, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26836767

RESUMO

Estradiol derived from neural aromatization of gonadal testosterone plays a key role in the perinatal organization of the neural circuitry underlying male sexual behavior. The aim of this study was to investigate the contribution of neural estrogen receptor (ER) ß in estradiol-induced effects without interfering with its peripheral functions. For this purpose, male mice lacking ERß in the nervous system were generated. Analyses of males in two consecutive tests with a time interval of two weeks showed an effect of experience, but not of genotype, on the latencies to the first mount, intromission, pelvic thrusting and ejaculation. Similarly, there was an effect of experience, but not of genotype, on the number of thrusts and mating length. Neural ERß deletion had no effect on the ability of males to adopt a lordosis posture in response to male mounts, after castration and priming with estradiol and progesterone. Indeed, only low percentages of both genotypes exhibited a low lordosis quotient. It also did not affect their olfactory preference. Quantification of tyrosine hydroxylase- and kisspeptin-immunoreactive neurons in the preoptic area showed unaffected sexual dimorphism of both populations in mutants. By contrast, the number of androgen receptor- and ERα-immunoreactive cells was significantly increased in the bed nucleus of stria terminalis of mutant males. These data show that neural ERß does not play a crucial role in the organization and activation of the neural circuitry underlying male sexual behavior. These discrepancies with the phenotype of global ERß knockout models are discussed.


Assuntos
Receptor beta de Estrogênio/genética , Camundongos , Mutagênese/genética , Gravidez , Comportamento Sexual Animal/fisiologia , Animais , Deleção Cromossômica , Feminino , Fertilidade/genética , Hipotálamo Anterior/metabolismo , Masculino , Camundongos Knockout , Neuroglia/metabolismo , Neurônios/metabolismo , Área Pré-Óptica/fisiologia , Núcleos Septais/metabolismo
11.
Sci Rep ; 5: 17457, 2015 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-26640081

RESUMO

Reproductive behavior and physiology in adulthood are controlled by hypothalamic sexually dimorphic neuronal networks which are organized under hormonal control during development. These organizing effects may be disturbed by endocrine disrupting chemicals (EDCs). To determine whether developmental exposure to Ethinylestradiol (EE2) may alter reproductive parameters in adult male mice and their progeny, Swiss mice (F1 generation) were exposed from prenatal to peripubertal periods to EE2 (0.1-1 µg/kg/d). Sexual behavior and reproductive physiology were evaluated on F1 males and their F2, F3 and F4 progeny. EE2-exposed F1 males and their F2 to F4 progeny exhibited EE2 dose-dependent increased sexual behavior, with reduced latencies of first mount and intromission, and higher frequencies of intromissions with a receptive female. The EE2 1 µg/kg/d exposed animals and their progeny had more calbindin immunoreactive cells in the medial preoptic area, known to be involved in the control of male sexual behavior in rodents. Despite neuroanatomical modifications in the Gonadotropin-Releasing Hormone neuron population of F1 males exposed to both doses of EE2, no major deleterious effects on reproductive physiology were detected. Therefore EE2 exposure during development may induce a hypermasculinization of the brain, illustrating how widespread exposure of animals and humans to EDCs can impact health and behaviors.


Assuntos
Etinilestradiol/farmacologia , Sistemas Neurossecretores/efeitos dos fármacos , Comportamento Sexual Animal/efeitos dos fármacos , Animais , Calbindinas/metabolismo , Feminino , Fertilidade/efeitos dos fármacos , Genitália/efeitos dos fármacos , Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Masculino , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Reprodução/efeitos dos fármacos , Testosterona/sangue
12.
Hum Mol Genet ; 24(25): 7326-38, 2015 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-26464488

RESUMO

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


Assuntos
Receptor beta de Estrogênio/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Animais , Estradiol/metabolismo , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/genética , Ciclo Estral/fisiologia , Feminino , Hormônio Liberador de Gonadotropina/genética , Hipotálamo/metabolismo , Kisspeptinas/genética , Camundongos , Puberdade/genética , Puberdade/metabolismo
13.
Theriogenology ; 84(6): 880-6, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26117287

RESUMO

In seasonally anovulatory goats, daily contact with photostimulated bucks for 15 days stimulates ovulations in most females. In this study, we determined whether ovulation could be induced in goats exposed to photostimulated bucks for less than 15 days. Bucks were rendered sexually active during the nonbreeding season by exposure to 2.5 months of long days from November 1. The control group of females was exposed to one photostimulated buck for 15 days (n = 12). Other three experimental groups were exposed to males (n = 1 per group) for 1, 5, or 10 days (n = 14 or 15 females per group). Ovulations were determined by measurement of daily plasma progesterone concentrations during 17 days. All females from the control and experimental groups ovulated at least once during the experiment (P > 0.05). Furthermore, the proportions of goats that displayed a short luteal phase followed by a new ovulation, or a normal luteal phase after being in contact with males, did not differ between groups depending on the duration of time of contact with the photostimulated males (P > 0.05). In contrast, the proportions of females that displayed a short luteal phase followed by anovulation were greater in goats in contact with males for 1 day than in those in contact with males for 10 and 15 days (P < 0.05), whereas they did not differ from females exposed to males for 5 days (P > 0.05). We conclude that 1 day of contact with sexually active males is long enough to stimulate the ovulatory activity in seasonally anovulatory goats. However, a significantly higher proportion of females exposed to males for 1 day did not ovulate again after showing a short luteal phase.


Assuntos
Anestro/fisiologia , Cabras/fisiologia , Indução da Ovulação/veterinária , Comportamento Sexual Animal , Animais , Feminino , Fase Luteal , Masculino , Ovulação/fisiologia , Indução da Ovulação/métodos , Progesterona/sangue , Reprodução/fisiologia , Reprodução/efeitos da radiação , Estações do Ano , Fatores de Tempo
14.
J Endocrinol ; 220(3): 375-88, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24403293

RESUMO

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


Assuntos
Compostos Benzidrílicos/efeitos adversos , Exposição Materna/efeitos adversos , Sistemas Neurossecretores/efeitos dos fármacos , Fenóis/efeitos adversos , Efeitos Tardios da Exposição Pré-Natal/psicologia , Animais , Estradiol/metabolismo , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Sistemas Neurossecretores/metabolismo , Ovário/crescimento & desenvolvimento , Ovário/metabolismo , Gravidez , Efeitos Tardios da Exposição Pré-Natal/etiologia , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Área Pré-Óptica/efeitos dos fármacos , Comportamento Sexual Animal/efeitos dos fármacos , Útero/crescimento & desenvolvimento , Útero/metabolismo
15.
Endocrinology ; 155(2): 502-12, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24265451

RESUMO

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


Assuntos
Compostos Benzidrílicos/administração & dosagem , Estrogênios não Esteroides/administração & dosagem , Rede Nervosa/efeitos dos fármacos , Fenóis/administração & dosagem , Área Pré-Óptica/efeitos dos fármacos , Comportamento Sexual Animal/efeitos dos fármacos , Administração Oral , Animais , Comportamento de Escolha/efeitos dos fármacos , Receptor alfa de Estrogênio/metabolismo , Comportamento Exploratório/efeitos dos fármacos , Kisspeptinas/metabolismo , Masculino , Camundongos , Rede Nervosa/metabolismo , Área Pré-Óptica/metabolismo , Receptores Androgênicos/metabolismo , Olfato/efeitos dos fármacos
16.
Gen Comp Endocrinol ; 188: 3-8, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23523710

RESUMO

Pheromones are known to trigger either short-term behavioral responses, usually referred to as "releaser effects", or more long-term physiological changes, known as "primer effects", which especially affect reproductive function at the level of the gonadotrope axis. The precise mechanisms through which pheromones interact with the gonadotrope axis in the hypothalamus is not fully known. We propose that the neuropeptide Kisspeptin, could be a specific target of primer pheromones, allowing these pheromones to modulate the gonadotrope axis and GnRH activity. This emerging hypothesis is discussed in the context of puberty acceleration in female mice and the male effect in female ungulates (sheep or goat). These examples have been chosen to illustrate the diversity of the reproductive contexts in mammals and potential mechanisms affected by primer effects at the level of the gonadotrope axis.


Assuntos
Kisspeptinas/metabolismo , Mamíferos/metabolismo , Mamíferos/fisiologia , Feromônios/metabolismo , Animais , Feminino , Hipotálamo/metabolismo , Masculino , Nervo Olfatório/metabolismo , Nervo Olfatório/fisiologia
17.
Physiol Behav ; 104(3): 392-7, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21557959

RESUMO

We assessed the development of individual discrimination resulting from direct contact/familiarization in ewes. Unfamiliar ewes were introduced during 6, 24 or 72 h in groups of already familiarized ewes. At the end of this contact period, the development of social recognition with the animal that has been introduced was assessed using two different types of tests: a Y-maze preference test and a delayed paired close encounters test where tested ewes are successively and randomly interacting with the familiar animal and an unfamiliar conspecific. The results of both tests showed that ewes developed a recognition of the familiar animal in comparison to a completely unfamiliar female. However, this preference was evidenced after 24 h of contact when using the paired close encounters test whereas it appears only after 72 h when using the Y-maze test, suggesting that the paired close encounters test is a more sensitive methodology to assess the development of social familiarization. The importance of estrogens, in the formation of social familiarization was also evaluated. To this end, social recognition in the paired close encounters test was compared between ovariectomized animals receiving estrogen implants or not. Despite significant high levels of estradiol in estrogen implanted females, no major differences in recognition appeared between groups, suggesting that in our conditions estrogens do not have major influence on social familiarization.


Assuntos
Comportamento Animal/fisiologia , Cabras/fisiologia , Cabras/psicologia , Reconhecimento Psicológico/fisiologia , Comportamento Social , Análise de Variância , Animais , Discriminação Psicológica/fisiologia , Estradiol/sangue , Estrogênios/administração & dosagem , Feminino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Bulbo Olfatório/efeitos dos fármacos , Bulbo Olfatório/fisiologia , Ovariectomia , Fatores de Tempo
18.
Horm Behav ; 57(4-5): 434-40, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20109458

RESUMO

In rodent species, sexual differentiation of the brain for many reproductive processes depends largely on estradiol. This was recently confirmed again by using the alpha-fetoprotein knockout (AFP-KO) mouse model, which lacks the protective actions of alpha-fetoprotein against maternal estradiol and as a result represents a good model to determine the contribution of prenatal estradiol to the sexual differentiation of the brain and behavior. Female AFP-KO mice were defeminized and masculinized with regard to their neuroendocrine responses as well as sexual behavior. Since parental behavior is also strongly sexually differentiated in mice, we used the AFP-KO mouse model here to ask whether parental responses are differentiated prenatally under the influence of estradiol. It was found that AFP-KO females showed longer latencies to retrieve pups to the nest and also exhibited lower levels of crouching over the pups in the nest in comparison to WT females. In fact, they resembled males (WT and AFP-KO). Other measures of maternal behavior, for example the incidence of infanticide, tended to be higher in AFP-KO females than in WT females but this increase failed to reach statistical significance. The deficits observed in parental behavior of AFP-KO females could not be explained by any changes in olfactory function, novelty recognition or anxiety. Thus our results suggest that prenatal estradiol defeminizes the parental brain in mice.


Assuntos
Estradiol/farmacologia , Comportamento Materno/fisiologia , Comportamento Paterno , alfa-Fetoproteínas/genética , alfa-Fetoproteínas/fisiologia , Animais , Ansiedade/psicologia , Feminino , Habituação Psicofisiológica/fisiologia , Masculino , Camundongos , Camundongos Knockout , Odorantes , Orquiectomia , Ovariectomia , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Reconhecimento Psicológico/fisiologia , Diferenciação Sexual , Olfato/fisiologia
19.
Behav Brain Res ; 194(1): 52-65, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18638505

RESUMO

In rats, expression of the immediate early gene, c-fos observed in the brain following male copulatory behavior relates mostly to the detection of olfactory information originating from the female and to somatosensory feedback from the penis. However, quail, like most birds, are generally considered to have a relatively poorly developed sense of smell. Furthermore, quail have no intromittent organ (e.g., penis). It is therefore intriguing that expression of male copulatory behavior induces in quail and rats a similar pattern of c-fos expression in the medial preoptic area (mPOA), bed nucleus of the stria terminalis (BSTM) and parts of the amygdala. We analyzed here by immunocytochemistry Fos expression in the mPOA/BSTM/amygdala of male quail that had been allowed to copulate with a female during standardized tests. Before these tests, some of the males had either their nostrils plugged, or their cloacal area anesthetized, or both. A control group was not exposed to females. These manipulations did not affect frequencies of male sexual behavior and all birds exposed to a female copulated normally. In the mPOA, the increased Fos expression induced by copulation was not affected by the cloacal gland anesthesia but was markedly reduced in subjects deprived of olfactory input. Both manipulations affected copulation-induced Fos expression in the BSTM. No change in Fos expression was observed in the amygdala. Thus immediate early gene expression in the mPOA and BSTM of quail is modulated at least in part by olfactory cues and/or somatosensory stimuli originating from the cloacal gland. Future work should specify the nature of these stimuli and their function in the expression of avian male sexual behavior.


Assuntos
Encéfalo/metabolismo , Transtornos do Olfato/complicações , Transtornos do Olfato/patologia , Proteínas Oncogênicas v-fos/metabolismo , Comportamento Sexual Animal/fisiologia , Glândulas Sudoríparas/fisiologia , Tonsila do Cerebelo/metabolismo , Análise de Variância , Anestésicos Locais/farmacologia , Animais , Comportamento Animal , Encéfalo/patologia , Contagem de Células/métodos , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Lidocaína/farmacologia , Masculino , Orquiectomia/métodos , Área Pré-Óptica/metabolismo , Codorniz , Núcleos Septais/metabolismo , Comportamento Sexual Animal/efeitos dos fármacos , Glândulas Sudoríparas/efeitos dos fármacos , Sistema Urinário/anatomia & histologia
20.
J Neurosci ; 27(24): 6563-72, 2007 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-17567817

RESUMO

Estrogens are classically viewed as hormones that bind to intracellular receptors, which then act as transcription factors to modulate gene expression; however, they also affect many aspects of neuronal functioning by rapid nongenomic actions. Brain estrogen production can be regulated within minutes by changes in aromatase (estrogen synthase) activity as a result of calcium-dependent phosphorylations of the enzyme. To determine the effects of rapid changes in estrogen availability on male copulatory behavior, we mimicked in male mice the rapid upregulation and downregulation of brain estrogen concentration that should occur after inactivation or activation of aromatase activity. A single injection of different aromatase inhibitors [Vorozole, 1,4,6-androstatrien-3,17-dione (ATD), or its metabolite 17-OH-ATD (1,4,6-androstatrien-17beta-ol-3-one)] almost completely suppressed male sexual behavior (mounts and intromissions) expressed 10-20 min later by C57BL/6J mice but did not affect behavior in aromatase knock-out (ArKO) mice, activated by daily injections of estradiol benzoate, thereby confirming the specificity of the behavioral inhibition observed in wild-type mice. The rapid ATD-induced inhibition was reversed by the simultaneous injection of a large dose of estradiol. A single injection of estradiol to ArKO mice also activated male sexual behavior within 15 min. Thus, rapid increases or decreases in brain estrogen concentrations are followed within minutes by corresponding changes in male sexual behavior. Sexual behavior can thus be used to monitor changes in local estrogen concentrations and analyze the mechanisms mediating the rapid decline in estrogen signaling that takes place after inhibition of estrogen synthesis.


Assuntos
Química Encefálica/fisiologia , Encéfalo/metabolismo , Estrogênios/metabolismo , Comportamento Sexual Animal/fisiologia , Animais , Aromatase/deficiência , Inibidores da Aromatase/farmacologia , Comportamento Animal , Encéfalo/efeitos dos fármacos , Química Encefálica/efeitos dos fármacos , Interações Medicamentosas , Estradiol/farmacologia , Comportamento Exploratório/efeitos dos fármacos , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Comportamento Sexual Animal/efeitos dos fármacos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA