Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 334
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Reprod Dev ; 70(2): 115-122, 2024 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-38346724

RESUMO

The NR4A nuclear receptor family (NR4As), encompassing NR4A1, NR4A2, and NR4A3, exerts pivotal roles in cellular processes through intricate expression patterns and interactions. Despite the influence of some NR4As on anterior pituitary functions regulated by the hypothalamus, their physiological expression patterns remain unclear. In our prior work, we demonstrated the specific upregulation of NR4A3 in the rat anterior pituitary gland during the proestrus afternoon, coinciding with a gonadotropin surge. In this study, we investigated changes in pituitary Nr4a gene expression throughout the estrous cycle in rats and a gonadotropin surge-induced model. Nr4a1 and Nr4a2 gene expression significantly increased during proestrus, aligning with previous observations for Nr4a3. Furthermore, prolactin gene expression increased sequentially with rising Nr4a gene expression, while thyroid-stimulating hormone beta gene expression remained stable. Immunohistochemistry revealed a widespread and differential distribution of NR4A proteins in the anterior pituitary, with NR4A1 and NR4A3 being particularly abundant in thyrotrophs, and NR4A2 in gonadotrophs. In estrogen-treated ovariectomized rats, elevated luteinizing hormone secretion corresponded to markedly upregulated expression of Nr4a1, Nr4a2, and Nr4a3. In gonadotroph and somatomammotroph cell lines, gonadotropin- and thyrotropin-releasing hormones transiently and dose-dependently increased the expression of Nr4a genes. These findings suggest that hypothalamic hormone secretion during proestrus may induce the parallel expression of pituitary Nr4a genes, potentially influencing the pituitary gene expression program related to endocrine functions before and after ovulation.


Assuntos
Adeno-Hipófise , Hipófise , Feminino , Ratos , Animais , Proestro/fisiologia , Hipófise/metabolismo , Adeno-Hipófise/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Gonadotropinas/metabolismo
2.
Croat Med J ; 61(4): 346-353, 2020 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-32881433

RESUMO

AIM: To determine the changes in skin temperature and brown adipose tissue (BAT) activity throughout the estrous cycle as well as the regularity of the estrous cycle in mice. METHODS: We assessed the differences in the duration of the estrous cycle and its phases between 3- and 8-month-old female mice (n=18). Skin temperature and BAT activity were measured by infrared technology and compared with human menstrual cycle. RESULTS: Young and old female mice did not differ significantly in the estrous cycle length. However, young animals had longer diestrus and shorter proestrus phase. In contrast with women, mice showed age-dependent changes in body temperature and BAT activity during the estrus cycle. CONCLUSION: Establishing the pattern of temperature and BAT activity changes could be used to determine the estrous cycle phase before performing experiments without disturbing the animal. However, since the regulation of BAT activity during the estrous cycle was age-dependent, very complex, and varied significantly from women, further studies are needed to develop a non-invasive method for determining the phase of the estrous cycle.


Assuntos
Tecido Adiposo Marrom/metabolismo , Ciclo Estral/fisiologia , Temperatura Cutânea/fisiologia , Termografia/métodos , Animais , Animais de Laboratório , Estro/fisiologia , Feminino , Raios Infravermelhos , Camundongos , Camundongos Endogâmicos C57BL , Proestro/fisiologia
3.
J Neurosci ; 38(5): 1061-1072, 2018 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-29114074

RESUMO

Estradiol feedback regulates gonadotropin-releasing hormone (GnRH) neurons and subsequent luteinizing hormone (LH) release. Estradiol acts via estrogen receptor α (ERα)-expressing afferents of GnRH neurons, including kisspeptin neurons in the anteroventral periventricular (AVPV) and arcuate nuclei, providing homeostatic feedback on episodic GnRH/LH release as well as positive feedback to control ovulation. Ionotropic glutamate receptors are important for estradiol feedback, but it is not known where they fit in the circuitry. Estradiol-negative feedback decreased glutamatergic transmission to AVPV and increased it to arcuate kisspeptin neurons; positive feedback had the opposite effect. Deletion of ERα in kisspeptin cells decreased glutamate transmission to AVPV neurons and markedly increased it to arcuate kisspeptin neurons, which also exhibited increased spontaneous firing rate. KERKO mice had increased LH pulse frequency, indicating loss of negative feedback. These observations indicate that ERα in kisspeptin cells is required for appropriate differential regulation of these neurons and neuroendocrine output by estradiol.SIGNIFICANCE STATEMENT The brain regulates fertility through gonadotropin-releasing hormone (GnRH) neurons. Ovarian estradiol regulates the pattern of GnRH (negative feedback) and initiates a surge of release that triggers ovulation (positive feedback). GnRH neurons do not express the estrogen receptor needed for feedback (estrogen receptor α [ERα]); kisspeptin neurons in the arcuate and anteroventral periventricular nuclei are postulated to mediate negative and positive feedback, respectively. Here we extend the network through which feedback is mediated by demonstrating that glutamatergic transmission to these kisspeptin populations is differentially regulated during the reproductive cycle and by estradiol. Electrophysiological and in vivo hormone profile experiments on kisspeptin-specific ERα knock-out mice demonstrate that ERα in kisspeptin cells is required for appropriate differential regulation of these neurons and for neuroendocrine output.


Assuntos
Estradiol/farmacologia , Glutamatos/fisiologia , Hipotálamo/citologia , Hipotálamo/fisiologia , Kisspeptinas/fisiologia , Neurônios/fisiologia , Receptores de Estrogênio/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/fisiologia , Dinorfinas/farmacologia , Feminino , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/fisiologia , Hipotálamo/efeitos dos fármacos , Hormônio Luteinizante/fisiologia , Camundongos , Núcleos da Linha Média do Tálamo/fisiologia , Neurônios/efeitos dos fármacos , Hipófise/efeitos dos fármacos , Hipófise/fisiologia , Proestro/fisiologia , Receptores Ionotrópicos de Glutamato/efeitos dos fármacos , Receptores Ionotrópicos de Glutamato/fisiologia , Transmissão Sináptica/efeitos dos fármacos , Receptor ERRalfa Relacionado ao Estrogênio
4.
J Cell Physiol ; 233(9): 6591-6603, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29115663

RESUMO

Annexin A1 (AnxA1) is a glucocorticoid-regulated anti-inflammatory protein secreted by phagocytes and other specialised cells. In the endocrine system, AnxA1 controls secretion of steroid hormones and it is abundantly expressed in the testis, ovaries, placenta and seminal fluid, yet its potential modulation of fertility has not been described. Here, we observed that AnxA1 knockout (KO) mice delivered a higher number of pups, with a higher percentage of female offsprings. This profile was not dependent on the male features, as sperm from KO male mice did not present functional alterations, and had an equal proportion of Y and X chromosomes, comparable to wild type (WT) male mice. Furthermore, mismatched matings of male WT mice with female KO yielded a higher percentage of female pups per litter, a phenomenon which was not observed when male KO mice mated with female WT animals. Indeed, AnxA1 KO female mice displayed several differences in parameters related to gestation including (i) an arrested estrous cycle at proestrus phase; (ii) increased sites of implantation; (iii) reduced pre- and post-implantation losses; (iv) exacerbated features of the inflammatory reaction in the uterine fluid during implantation phase; and (v) enhanced plasma progesterone in the beginning of pregnancy. In summary, herein we highlight that AnxA1 pathway as a novel determinant of fundamental non-redundant regulatory functions during early pregnancy.


Assuntos
Anexina A1/metabolismo , Implantação do Embrião/fisiologia , Animais , Ciclo Estral/metabolismo , Ciclo Estral/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Modelos Animais , Gravidez , Proestro/metabolismo , Proestro/fisiologia , Razão de Masculinidade , Útero/metabolismo , Útero/fisiologia , Cromossomo X/metabolismo , Cromossomo X/fisiologia , Cromossomo Y/metabolismo , Cromossomo Y/fisiologia
5.
Reprod Fertil Dev ; 30(11): 1541-1552, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29778102

RESUMO

The aim of the present study was to investigate the effects of a strategy for extending pro-oestrus (the interval between luteolysis and ovulation) in an oestrus synchronisation protocol (named J-Synch) in beef heifers on follicular growth, sexual steroid concentrations, the oestrogen receptor ERα and progesterone receptors (PR) in the uterus, insulin-like growth factor (IGF) 1 and pregnancy rates. In Experiment 1, heifers treated with the new J-Synch protocol had a longer pro-oestrus period than those treated with the conventional protocol (mean (±s.e.m.) 93.7±12.9 vs 65.0±13.7h respectively; P<0.05). The rate of dominant follicle growth from the time of progesterone device removal to ovulation was greater in heifers in the J-Synch than conventional group (P<0.05). Luteal area and serum progesterone concentrations were greater in the J-Synch Group (P<0.05) for the 12 days after ovulation. Progesterone receptor (PGR) staining on Day 6 after ovulation in the uterine stroma was lower in the J-Synch than conventional group (P<0.05), and the expression of PR gene (PGR) and IGF1 gene tended to be lower in J-Synch-treated heifers (P<0.1). In Experiment 2 (n=2349), the pregnancy rate 30-35 days after fixed-time AI (FTAI) was greater for heifers in the J-Synch than conventional group (56.1% vs 50.7% respectively). In conclusion, our strategy for extending pro-oestrus (i.e. the J-Synch protocol) significantly improves pregnancy establishment in beef heifers. This improvement was related to an increased rate of growth of the dominant ovulatory follicle, greater progesterone concentrations during the ensuing luteal phase and different uterine patterns of PGR and IGF1, which may have favoured embryo development and pregnancy establishment.


Assuntos
Estradiol/análogos & derivados , Sincronização do Estro/fisiologia , Ovário/fisiologia , Proestro/fisiologia , Progesterona/administração & dosagem , Útero/fisiologia , Animais , Bovinos , Estradiol/administração & dosagem , Estradiol/sangue , Sincronização do Estro/efeitos dos fármacos , Feminino , Folículo Ovariano/diagnóstico por imagem , Folículo Ovariano/efeitos dos fármacos , Ovário/diagnóstico por imagem , Ovário/efeitos dos fármacos , Gravidez , Proestro/efeitos dos fármacos , Progesterona/sangue , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Útero/diagnóstico por imagem , Útero/efeitos dos fármacos
6.
Hippocampus ; 26(1): 110-7, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26190171

RESUMO

When circulating 17ß estradiol (E2) is elevated to proestrous levels, hippocampus-dependent learning and memory is enhanced in female rodents, nonhuman primates, and women due to heightened synaptic function at hippocampal synapses. We previously reported that proestrous-like levels of E2 administered to young adult ovariectomized (OVX) female rats increases the magnitude of LTP at CA3 Schaffer collateral (SC)-CA1 synapses only when dendritic spine density, the NMDAR/AMPAR ratio, and current mediated by GluN2B-containing NMDA receptors (NMDARs) are simultaneously increased. We also reported that this increase in GluN2B-mediated NMDAR current in area CA1 is causally related to the E2-induced increase in novel object recognition, tying together heightened synaptic function with improved learning and memory. In addition to SC inputs, innervation from the entorhinal cortex in the temporoammonic (TA) pathway onto CA1 distal dendrites in stratum lacunosum-moleculare is critical for spatial memory formation and retrieval. It is not known whether E2 modulates TA-CA1 synapses similarly to SC-CA1 synapses. Here, we report that 24 hours post-E2 injection, dendritic spine density on CA1 pyramidal cell distal dendrites and current mediated by GluN2B-containing NMDARs at TA-CA1 synapses is increased, similarly to our previous findings at SC-CA1 synapses. However, in contrast to SC-CA1 synapses, AMPAR transmission at TA-CA1 synapses is significantly increased, and there is no effect on the LTP magnitude. Pharmacological blockade of GluN2B-containing NMDARs or ERK activation, which occurs downstream of synaptic but not extrasynaptic GluN2B-containing NMDARs, attenuates the LTP magnitude only in slices from E2-treated rats. These data show that E2 recruits a causal role for GluN2B-containing NMDARs and ERK signaling in the induction of LTP, cellular mechanisms not required for LTP induction at TA-CA1 synapses in vehicle-treated OVX female rats.


Assuntos
Região CA1 Hipocampal/efeitos dos fármacos , Estradiol/farmacologia , Estrogênios/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Potenciação de Longa Duração/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/fisiologia , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Feminino , Potenciação de Longa Duração/fisiologia , Vias Neurais/citologia , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiologia , Ovariectomia , Proestro/fisiologia , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Sinapses/efeitos dos fármacos , Sinapses/fisiologia , Lobo Temporal/citologia , Lobo Temporal/efeitos dos fármacos , Lobo Temporal/fisiologia , Técnicas de Cultura de Tecidos , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/metabolismo
7.
Am J Physiol Regul Integr Comp Physiol ; 305(3): R271-80, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23678032

RESUMO

Ovarian hormones are thought to modulate sleep and fluctuations in the hormonal milieu are coincident with sleep complaints in women. In female rats, estradiol increases waking and suppresses sleep. In this study, we asked whether this effect is mediated via circadian or homeostatic regulatory mechanisms. Ovariectomized female rats received daily injections of estradiol benzoate (EB) or sesame oil that mimicked the rapid increase and subsequent decline of circulating estradiol at proestrus. In one experiment, animals were sleep deprived for 6 h starting at lights-on, so that recovery began in the mid-light phase; in the second experiment, animals were sleep deprived starting in the mid-light phase, so that recovery began at lights-off. EB suppressed baseline rapid eye movement (REM) and non-REM (NREM) sleep and increased waking in the dark phase. In both experiments, EB enhanced REM recovery in the light phase while suppressing it in the dark compared with oil; this effect was most pronounced in the first 6 h of recovery. By contrast, NREM recovery was largely unaffected by EB. In summary, EB enhanced waking and suppressed sleep, particularly REM sleep, in the dark under baseline and recovery conditions. These strong temporally dependent effects suggest that EB consolidates circadian sleep-wake rhythms in female rats.


Assuntos
Ritmo Circadiano/fisiologia , Estradiol/farmacologia , Sono REM/efeitos dos fármacos , Animais , Escuridão , Interpretação Estatística de Dados , Eletrodos Implantados , Eletroencefalografia , Eletromiografia , Estradiol/sangue , Feminino , Luz , Ovariectomia , Proestro/fisiologia , Ratos , Ratos Sprague-Dawley , Sono/efeitos dos fármacos , Privação do Sono/tratamento farmacológico , Privação do Sono/fisiopatologia , Fases do Sono/efeitos dos fármacos , Vigília/efeitos dos fármacos
8.
Sci Rep ; 13(1): 20988, 2023 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-38017045

RESUMO

Despite the prevalent expression of freezing behavior following Pavlovian fear conditioning, a growing body of literature suggests potential sex differences in defensive responses. Our study investigated how female defensive behaviors are expressed in different threat situations and modulated by the estrous cycle. We aimed to compare freezing and flight-like responses during the acquisition and retrieval of fear conditioning using two distinct unconditioned stimuli (US) in two different spatial configurations: (1) electrical footshock (FUS) in a small, conventional enclosure with a grid floor, and (2) a predator-like robot (PUS) in a spacious, open arena. Fear conditioning with FUS showed no substantial differences between male and female rats of two different estrous cycles (proestrus and diestrus) in the levels of freezing and flight. However, when PUS was employed, proestrus female rats showed significantly more flight responses to the CS during both acquisition and the retrieval compared to the male and diestrus female rats. Taken together, our findings suggest that hormonal influences on the choice of defensive strategies in threat situations are significantly modulated by both the type of US and the spatial configuration of the environment.


Assuntos
Condicionamento Clássico , Ciclo Estral , Ratos , Feminino , Masculino , Animais , Ciclo Estral/fisiologia , Medo/fisiologia , Proestro/fisiologia , Comportamento Animal/fisiologia
9.
J Dairy Sci ; 95(5): 2513-22, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22541478

RESUMO

The objectives were to compare the effects of 2 methods of presynchronization and 2 lengths of proestrus on fertility of grazing dairy cows subjected to a 5-d timed artificial insemination (AI) protocol at initiation of breeding season. Lactating dairy cows (n=1,754) from 3 seasonal grazing farms were blocked within farm by breed, parity, and days in milk (DIM). Study d 0 was considered the day of AI of cows in COS72 (72h of proestrus). Within each block, cows were randomly assigned to 1 of 2 presynchronization treatments: a PGF(2α)-based program, Presynch, consisting of 2 injections of PGF(2α) administered on d -32 and -18, or a PGF(2α)-GnRH-based program, Double-Ovsynch (DO), consisting of GnRH on d -25, PGF(2α) on d -18, and GnRH on d -15. Within each of the 2 presynchronization treatments, cows were randomly assigned to 1 of 2 lengths of proestrus within the 5-d timed AI protocol, consisting of GnRH on d -8, PGF(2α) on d -3 and -2, and GnRH+AI at either 58 h (COS58) or 72 h (COS72) after the d -3 PGF(2α) injection. Ovaries were scanned by ultrasonography twice, on d -42 and -32, to determine estrous cyclicity before enrollment in the study. Blood was sampled and analyzed for concentrations of estradiol on the day of AI. Pregnancies per AI (P/AI) were determined 30 and 65 d after AI. Presynchronization did not affect the concentration of estradiol at AI (DO=6.4 vs. Presynch=5.8 pg/mL), detection of estrus at AI (20.8 vs. 25.9%), or P/AI on d 30 (56.8 vs. 59.1%) and 65 (52.5 vs. 52.4%) after the first AI. Cows receiving COS72 had increased concentration of estradiol (6.6 vs. 5.5 pg/mL) and detection of estrus at AI (28.5 vs. 10.8%) compared with cows receiving COS58. Length of proestrus did not affect P/AI on d 30 (COS72=58.7 vs. COS58=56.1%) but, in Presynch cows, COS58 was detrimental to fertility on d 65 after AI (54.9 vs. 46.5%). Pregnancy loss between gestational d 30 and 65 was greater for Presynch than for DO (7.6 vs. 11.3%), but it was not affected by length of proestrus. Estrous cyclic cows had greater P/AI than anovular cows on d 30 (61.7 vs. 35.1%) and 65 (56.1 vs. 30.7%), but no interaction between estrous cyclic status and treatments was detected. Crossbred Holstein/Jersey cows had superior fertility than their purebred counterparts during the breeding season. The Presynch and DO protocols resulted in similar fertility with no overall difference between the presynchronization methods; however, limiting the length of proestrus to 58 h reduced P/AI in the 5-d timed AI protocol when cows had their estrous cycle presynchronized with Presynch but not with DO.


Assuntos
Sincronização do Estro/métodos , Inseminação Artificial/veterinária , Proestro/fisiologia , Animais , Bovinos , Indústria de Laticínios/métodos , Estradiol/sangue , Feminino , Fertilidade/fisiologia , Inseminação Artificial/métodos , Gravidez , Fatores de Tempo
10.
Eksp Klin Farmakol ; 75(2): 3-6, 2012.
Artigo em Russo | MEDLINE | ID: mdl-22550850

RESUMO

Results of a comparative study of the effects of chronic administration of the D2-receptor agonist quinperole (0.1 mg/kg, i.p.) and the D2-receptor antagonist sulpiride (10.0 mg/kg, i.p.) for 14 days on anxiety- and depressive-like behavior in key phases of the ovarian cycle in adult female rats are presented. The model of depression in rats was implemented in Porsolt test, while the anxiety level was assessed in the elevated plus maze test. It is established that the chronic administration of quinperole produced an anxiolytic action in female rats during diesrous, estrous and proestrous phases, but failed to modify depression-like behavior during the entire ovarian cycle. Sulpiride administration resulted in anxiogenic effect in all phases of the ovarian cycle. It was also found that sulpiride produced some modulation of depression-like behavior in connection to ovarian cycle phases, which was a prodepressive action at a moderate level of estrogens and an antidepressant effect at a reduced/enhanced level of estrogen. It is suggested that the extent of involvement of D2-receptors in the mechanisms of anxiety-depressive-like behavior can vary depending on alterations of the hormonal balance during the ovarian cycle. The data obtained are indicative of a close interaction between ovarian hormonal and dopaminergic systems of the brain involved in the mechanisms of anxiety and depression.


Assuntos
Ansiedade/psicologia , Depressão/psicologia , Ciclo Menstrual/psicologia , Quimpirol/farmacologia , Sulpirida/farmacologia , Adulto , Animais , Antidepressivos/farmacologia , Ansiedade/tratamento farmacológico , Ansiedade/fisiopatologia , Depressão/tratamento farmacológico , Depressão/fisiopatologia , Diestro/efeitos dos fármacos , Diestro/fisiologia , Dopamina/metabolismo , Agonistas de Dopamina/farmacologia , Antagonistas dos Receptores de Dopamina D2 , Estro/efeitos dos fármacos , Estro/fisiologia , Feminino , Humanos , Aprendizagem em Labirinto/efeitos dos fármacos , Ciclo Menstrual/efeitos dos fármacos , Ciclo Menstrual/fisiologia , Proestro/efeitos dos fármacos , Proestro/fisiologia , Ratos , Ratos Wistar , Receptores de Dopamina D2/agonistas
11.
Behav Neurosci ; 136(5): 349-363, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35254840

RESUMO

The ability to remember sequences of events is fundamental to episodic memory. While rodent studies have examined sex and estrous cycle in episodic-like spatial memory tasks, little is known about these biological variables in memory for sequences of events that depend on representations of temporal context. We investigated the role of sex and estrous cycle in rats during training and testing stages of a cross-species validated sequence memory task (Jayachandran et al., 2019). Rats were trained on a two four-odor sequence memory task delivered on opposite ends of a linear track. Training occurred in six successive stages starting with learning to poke in a nose-port for ≥ 1.2 s; eventually demonstrating sequence memory by holding their nose in the port ≥ 1 s for in-sequence odors and < 1 s for out-of-sequence odors. Performance was analyzed across sex and estrous cycle (proestrus, estrus, metestrus, and diestrus), the latter being determined by cellular composition of a daily vaginal lavage. We found no evidence of sex differences in asymptotic sequence memory performance, similar to humans performing an analogous task (Reeders et al., 2021). Likewise, no differences in sequence memory performance were found across the estrous cycle. Some caveats are that males acquired out-of-sequence trials faster during training with a 3-odor sequence, but this apparent advantage did not carry over to the 4-odor sequence. Additionally, males had shorter poke times overall which seem consistent with a decreased overall response inhibition because they occurred regardless of sequence demands. Together, these results suggest sex and estrous cycle are not major factors in sequence memory capacities. (PsycInfo Database Record (c) 2022 APA, all rights reserved).


Assuntos
Ciclo Estral , Estro , Animais , Ciclo Estral/fisiologia , Estro/fisiologia , Feminino , Humanos , Masculino , Metestro/fisiologia , Proestro/fisiologia , Ratos , Memória Espacial
12.
Toxicol Pathol ; 39(4): 641-52, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21551027

RESUMO

Evaluation of ovarian toxicity requires an understanding of the physiological changes related to the estrous cycle in the ovary. The authors investigated the transitional gene expression profile of ovulatory follicles in rats that show normal estrous cyclicity. Ovaries were collected at 10:00 and 22:00 on the proestrus day and at 10:00 on the estrus day. Ovarian follicles or early corpora lutea were isolated using laser microdissection, and extracted total RNA was analyzed using microarray technology. Clustering analysis revealed four different expression patterns: transient up- or down-regulation only at 22:00 on the proestrus day (pattern 1), up- or down-regulation only at 10:00 on the estrus day (pattern 2), continuous increase at 22:00 on the proestrus day and at 10:00 on the estrus day (pattern 3), and up- or down-regulation at 22:00 on the proestrus day and level maintenance at 10:00 on the estrus day (pattern 4). In addition, these probe sets were functionally categorized in each pattern using the Ingenuity Pathways Analysis database. These data will aid in understanding the physiology of ovulation and may be useful in assessing ovarian toxicity and its mechanism, such as in investigations of chemical-induced ovulatory impairment.


Assuntos
Perfilação da Expressão Gênica/métodos , Folículo Ovariano/metabolismo , Ovulação/genética , Animais , Análise por Conglomerados , Corpo Lúteo/fisiologia , Regulação para Baixo , Estro/fisiologia , Feminino , Microdissecção , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Proestro/fisiologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Regulação para Cima
13.
Psychopharmacology (Berl) ; 238(4): 969-978, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33404736

RESUMO

RATIONALE: Heroin intake decreases during the proestrus phase of the estrous cycle in female rats. Circulating concentrations of both estradiol and progesterone peak during proestrus, and it is not known which of these hormones, or their combination, are responsible for these effects. OBJECTIVES: The purpose of this study was to determine the effects of estradiol, progesterone, and their combination on heroin self-administration in female rats. METHODS: In Experiment 1, the estrous cycle of intact female rats was tracked daily. If a rat was in proestrus, either the estrogen receptor antagonist, raloxifene, the progesterone receptor antagonist, mifepristone, or their combination was administered 30 min prior to a heroin self-administration session. In Experiment 2, separate groups of ovariectomized female rats were treated chronically with exogenous estradiol, progesterone, estradiol + progesterone, or vehicle, and heroin intake was examined over a 100-fold dose range. RESULTS: In Experiment 1, raloxifene, but not mifepristone, significantly blocked proestrus-associated decreases in heroin intake. In Experiment 2, estrogentreated rats self-administered less heroin than any other group and significantly less heroin than rats treated with progesterone. CONCLUSIONS: These data suggest that (1) estradiol but not progesterone is responsible for proestrus-associated decreases in heroin intake and (2) estradiol decreases heroin intake relative to progesterone. These data differ from those reported previously with stimulants and suggest that estrogen-based pharmacotherapies may be of value to women with opioid use disorder.


Assuntos
Estradiol/administração & dosagem , Heroína/administração & dosagem , Progesterona/administração & dosagem , Animais , Estradiol/farmacologia , Estro/fisiologia , Feminino , Heroína/farmacologia , Mifepristona/administração & dosagem , Mifepristona/farmacologia , Proestro/fisiologia , Progesterona/farmacologia , Ratos , Ratos Long-Evans
14.
Endocrinology ; 162(6)2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33824970

RESUMO

Coordination of ovulation and behavior is critical to reproductive success in many species. During the female estrous cycle, the preovulatory gonadotropin surge occurs when ovarian follicles reach maturity and, in rodents, it begins just before the daily onset of activity, ensuring that ovulation coincides with sex behavior. Timing of the surge relies on projections from the suprachiasmatic nucleus (SCN), the locus of the central circadian clock, to hypothalamic circuits that regulate gonadotropin secretion. The cellular mechanisms through which the SCN controls these circuits and gates the preovulatory surge to the appropriate estrous cycle stage, however, are poorly understood. We investigated in mice the functional impact of SCN arginine-vasopressin (AVP) neuron projections to kisspeptin (Kiss1) neurons in the rostral periventricular area of the third ventricle (RP3VKiss1), responsible for generating the preovulatory surge. Conditional anterograde tracing revealed that SCNAVP neurons innervate approximately half of the RP3VKiss1 neurons. Optogenetic activation of SCNAVP projections in brain slices caused an AVP-mediated stimulation of RP3VKiss1 action potential firing in proestrus, the cycle stage when the surge is generated. This effect was less prominent in diestrus, the preceding cycle stage, and absent in estrus, following ovulation. Remarkably, in estrus, activation of SCNAVP projections resulted in GABA-mediated inhibition of RP3VKiss1 neuron firing, an effect rarely encountered in other cycle stages. Together, these data reveal functional plasticity in SCNAVP neuron output that drives opposing effects on RP3VKiss1 neuron activity across the ovulatory cycle. This might contribute to gating activation of the preovulatory surge to the appropriate estrous cycle stage.


Assuntos
Relógios Circadianos/fisiologia , Ciclo Estral/fisiologia , Neurônios/fisiologia , Potenciais de Ação/fisiologia , Animais , Arginina Vasopressina/genética , Arginina Vasopressina/metabolismo , Mapeamento Encefálico , Plasticidade Celular/fisiologia , Ritmo Circadiano/fisiologia , Feminino , Kisspeptinas/metabolismo , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Proestro/fisiologia , Núcleo Supraquiasmático/metabolismo , Núcleo Supraquiasmático/fisiologia
15.
Stress ; 13(3): 269-75, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20392197

RESUMO

The temporomandibular joint (TMJ) formalin test was used to evaluate the effects of acute restraint stress on the nociceptive behavioral responses of female rats during proestrus and estrus phases of the estrous cycle. Rats were subjected to one session of restraint stress (15, 30 min or 1 h). They were then either immediately killed to allow the collection of blood for hormonal radioimmunoassay determinations or subjected to TMJ formalin test to evaluate nociception. All stress protocols significantly raised the plasma concentrations of corticosterone. The performance of rats subjected to 15 and 30 min of restraint stress was similar to that of control rats, whereas rats that were stressed for 1 h showed a decrease in nociceptive responses, during both proestrus and estrus phases. The stress-induced analgesia (SIA) was greater in the proestrus phase. To evaluate the role of kappa-opioid receptors, the selective receptor kappa-opioid antagonist nor-binaltorphimine (nor-BNI; 200 microg or saline) was injected into the TMJ 24 h prior to the 1 h stress period and the TMJ formalin test. The local administration of nor-BNI partially reversed the SIA during the proestrus phase. These findings suggest that (1) acute stress for 1 h can produce analgesia both during proestrus and estrus phases; this effect is greater during the proestrus phase and (2) kappa-opioid receptor activation is involved in the SIA observed in the proestrus phase.


Assuntos
Nociceptores/fisiologia , Medição da Dor , Dor/fisiopatologia , Estresse Fisiológico/fisiologia , Transtornos da Articulação Temporomandibular/fisiopatologia , Doença Aguda , Animais , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Estro/fisiologia , Feminino , Formaldeído/farmacologia , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Nociceptores/efeitos dos fármacos , Dor/induzido quimicamente , Proestro/fisiologia , Ratos , Ratos Wistar , Receptores Opioides/fisiologia , Restrição Física , Articulação Temporomandibular/efeitos dos fármacos , Articulação Temporomandibular/fisiologia , Transtornos da Articulação Temporomandibular/induzido quimicamente
16.
Horm Behav ; 57(2): 216-21, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19958772

RESUMO

The aim of this investigation was to evaluate overall DNA damage induced by experimental paradoxical sleep deprivation (PSD) in estrous-cycling and ovariectomized female rats to examine possible hormonal involvement during DNA damage. Intact rats in different phases of the estrous cycle (proestrus, estrus, and diestrus) or ovariectomized female Wistar rats were subjected to PSD by the single platform technique for 96 h or were maintained for the equivalent period as controls in home-cages. After this period, peripheral blood and tissues (brain, liver, and heart) were collected to evaluate genetic damage using the single cell gel (comet) assay. The results showed that PSD caused extensive genotoxic effects in brain cells, as evident by increased DNA migration rates in rats exposed to PSD for 96 h when compared to negative control. This was observed for all phases of the estrous cycle indistinctly. In ovariectomized rats, PSD also led to DNA damage in brain cells. No significant statistically differences were detected in peripheral blood, the liver or heart for all groups analyzed. In conclusion, our data are consistent with the notion that genetic damage in the form of DNA breakage in brain cells induced by sleep deprivation overrides the effects related to endogenous female sex hormones.


Assuntos
Dano ao DNA , Hormônios Esteroides Gonadais/metabolismo , Privação do Sono/genética , Privação do Sono/metabolismo , Sono REM , Animais , Sangue/metabolismo , Encéfalo/metabolismo , Corticosterona/metabolismo , Diestro/genética , Diestro/fisiologia , Estro/genética , Estro/fisiologia , Feminino , Fígado/metabolismo , Miocárdio/metabolismo , Ovariectomia , Proestro/genética , Proestro/fisiologia , Progesterona/metabolismo , Distribuição Aleatória , Ratos , Ratos Wistar , Testosterona/metabolismo
17.
J Am Assoc Lab Anim Sci ; 59(3): 282-287, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32241334

RESUMO

Vaginal cytology is the most common method of monitoring the estrous cycle in rats; however, this test requires specific technical training and can be subject to interpretation. Vaginal impedance offers a quicker and less technically challenging alternative and has been used successfully to identify estrus in normally cycling breeder rats. We hypothesize that vaginal impedance can also be used to stage the estrous cycle in rats that have been given luteinizing hormone releasing hormone (LHRH) for timed mating. Vaginal impedance measurements and vaginal cytology were performed in LHRH-primed female rats (n = 36) at the expected peak of proestrus and paired with proven stud males. Breeding success was determined by gross necropsy to detect embryo implantation sites in the female rats. We found that the predictive rates of vaginal cytology and impedance measurement for proestrus were similar; however, both methods resulted in high proportions of false positive and false negative determinations (28% and 31%, respectively). We further hypothesized that females respond to LHRH at variable rates, resulting in variable times of peak proestrus. To test this, vaginal impedance measurements were performed multiple times throughout the expected day of proestrus in LHRH-primed female rats (n = 36). Females were either paired with a male 24 h after reaching the proestrus threshold (n = 18) or paired according to our standard protocol at 1300 h on the day after the expected proestrus (n = 18). Sequential measurements reduced false positive and negative rates (14% and 8%, respectively). Pregnancy rates did not differ based on the time of pairing during expected estrus. Overall, we determined vaginal impedance can be more successful than vaginal cytology at identifying proestrus in the rat, but only if multiple measurements are taken.


Assuntos
Ciclo Estral , Ratos/fisiologia , Técnicas Reprodutivas/veterinária , Vagina/fisiologia , Técnicos em Manejo de Animais , Animais , Impedância Elétrica , Feminino , Hormônio Luteinizante/agonistas , Masculino , Gravidez , Proestro/fisiologia
18.
Neuroscience ; 159(1): 7-15, 2009 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-19162130

RESUMO

The difference in visual object recognition by males and females suggests a sex-specific function in the medial prefrontal cortex (mPFC). In the present study, we performed an in vivo microdialysis study in three groups of rats (males, diestrous females, and proestrous females) to examine the potential sex difference in acetylcholine (ACh) release in the mPFC. The dialysate was automatically collected from the mPFC every 20 min for 24 h under freely moving conditions and the spontaneous locomotor activity was simultaneously monitored. Although ACh release in the mPFC during the dark phase was significantly greater than during the light phase in both sexes, the female rats consistently exhibited a significantly greater mean ACh release than the males. Spontaneous locomotor activity during the dark phase was also significantly greater than during the light phase in both sexes, but the females exhibited significantly greater spontaneous locomotor activity than the males. In addition, both sexes of rats were found to have significant positive correlations between ACh release and spontaneous locomotor activity, but females were found to have significantly greater correlation coefficients than males. Stereological methods were used to examine the number of choline acetyltransferase immunoreactive cells in the nucleus basalis magnocellularis and the horizontal diagonal band of Broca. The number of choline acetyltransferase immunoreactive cells in the nucleus basalis magnocellularis was also greater in females than males, suggesting a contribution to the higher ACh release in females. In contrast, no sex difference in the choline acetyltransferase immunoreactive cells was observed in the horizontal diagonal band of Broca. This is the first report to show a sex difference in the 24-h ACh release profile in the mPFC of behaving rats.


Assuntos
Acetilcolina/metabolismo , Ritmo Circadiano/fisiologia , Atividade Motora/fisiologia , Córtex Pré-Frontal/metabolismo , Caracteres Sexuais , Análise de Variância , Animais , Núcleo Basal de Meynert/metabolismo , Colina O-Acetiltransferase/metabolismo , Feixe Diagonal de Broca/metabolismo , Diestro/fisiologia , Feminino , Masculino , Microdiálise , Proestro/fisiologia , Ratos , Ratos Wistar , Vigília
19.
Reprod Biol Endocrinol ; 7: 60, 2009 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-19500372

RESUMO

BACKGROUND: Remodeling of the extracellular matrix is one of the most striking features observed in the uterus during the estrous cycle and after hormone replacement. Versican (VER) is a hyaluronan-binding proteoglycan that undergoes RNA alternative splicing, generating four distinct isoforms. This study analyzed the synthesis and distribution of VER in mouse uterine tissues during the estrous cycle, in ovariectomized (OVX) animals and after 17beta-estradiol (E2) and medroxyprogesterone (MPA) treatments, either alone or in combination. METHODS: Uteri from mice in all phases of the estrous cycle, and animals subjected to ovariectomy and hormone replacement were collected for immunoperoxidase staining for versican, as well as PCR and quantitative Real Time PCR. RESULTS: In diestrus and proestrus, VER was exclusively expressed in the endometrial stroma. In estrus and metaestrus, VER was present in both endometrial stroma and myometrium. In OVX mice, VER immunoreaction was abolished in all uterine tissues. VER expression was restored by E2, MPA and E2+MPA treatments. Real Time PCR analysis showed that VER expression increases considerably in the MPA-treated group. Analysis of mRNA identified isoforms V0, V1 and V3 in the mouse uterus. CONCLUSION: These results show that the expression of versican in uterine tissues is modulated by ovarian steroid hormones, in a tissue-specific manner. VER is induced in the myometrium exclusively by E2, whereas MPA induces VER deposition only in the endometrial stroma.


Assuntos
Ciclo Estral/fisiologia , Útero/fisiologia , Versicanas/genética , Versicanas/metabolismo , Animais , Diestro/fisiologia , Estrogênios/metabolismo , Estrogênios/farmacologia , Estro/fisiologia , Feminino , Técnicas Imunoenzimáticas , Medroxiprogesterona/metabolismo , Medroxiprogesterona/farmacologia , Camundongos , Proestro/fisiologia , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Útero/efeitos dos fármacos
20.
Neuroendocrinology ; 90(3): 292-300, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19684383

RESUMO

BACKGROUND: Estrogens are recognized as acting as modulators of pituitary cell renewal, sensitizing cells to mitogenic and apoptotic signals, thus participating in anterior pituitary homeostasis during the estrous cycle. The balance of pro- and antiapoptotic proteins of the Bcl-2 family is known to regulate cell survival and apoptosis. AIMS: In order to understand the mechanisms underlying apoptosis during the estrous cycle, we evaluated the expression of the proapoptotic protein Bax and the antiapoptotic proteins Bcl-2 and Bcl-xL in the anterior pituitary gland in cycling female rats as well as the influence of estradiol on the expression of these proteins in anterior pituitary cells of ovariectomized rats. METHODS/RESULTS: As determined by Western blot, the expression of Bax was higher in anterior pituitary glands from rats at proestrus than at diestrus I, Bcl-2 protein levels showed no difference and Bcl-xL expression was lower, thus increasing the Bax/Bcl-2 ratio at proestrus. Assessed by annexin V binding and flow cytometry, the percentage of apoptotic anterior pituitary cells was higher in rats at proestrus than at diestrus I. Chronic estrogen treatment in ovariectomized rats enhanced the Bax/Bcl-2 ratio and induced apoptosis. Moreover, incubation of cultured anterior pituitary cells from ovariectomized rats with 17beta-estradiol for 24 h increased the Bax/Bcl-2 ratio, decreased Bcl-xL expression and induced apoptosis. CONCLUSION: Our results demonstrate that estradiol increases the ratio between proapoptotic and antiapoptotic proteins of the Bcl-2 family. This effect could participate in the sensitizing action of estrogens to proapoptotic stimuli and therefore be involved in the high apoptotic rate observed at proestrus in the anterior pituitary gland.


Assuntos
Apoptose/fisiologia , Estradiol/metabolismo , Ciclo Estral/fisiologia , Adeno-Hipófise/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína X Associada a bcl-2/metabolismo , Animais , Anexina A5/metabolismo , Western Blotting , Contagem de Células , Diestro/fisiologia , Estrogênios/metabolismo , Feminino , Citometria de Fluxo , Ovariectomia , Proestro/fisiologia , Ratos , Ratos Wistar , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA