Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 335
Filter
1.
J Reprod Dev ; 70(2): 115-122, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38346724

ABSTRACT

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.


Subject(s)
Pituitary Gland, Anterior , Pituitary Gland , Female , Rats , Animals , Proestrus/physiology , Pituitary Gland/metabolism , Pituitary Gland, Anterior/metabolism , Gonadotropin-Releasing Hormone/metabolism , Gonadotropins/metabolism
2.
Sci Rep ; 13(1): 20988, 2023 11 28.
Article in English | MEDLINE | ID: mdl-38017045

ABSTRACT

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.


Subject(s)
Conditioning, Classical , Estrous Cycle , Rats , Female , Male , Animals , Estrous Cycle/physiology , Fear/physiology , Proestrus/physiology , Behavior, Animal/physiology
3.
Behav Neurosci ; 136(5): 349-363, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35254840

ABSTRACT

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).


Subject(s)
Estrous Cycle , Estrus , Animals , Estrous Cycle/physiology , Estrus/physiology , Female , Humans , Male , Metestrus/physiology , Proestrus/physiology , Rats , Spatial Memory
4.
Endocrinology ; 162(6)2021 06 01.
Article in English | MEDLINE | ID: mdl-33824970

ABSTRACT

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.


Subject(s)
Circadian Clocks/physiology , Estrous Cycle/physiology , Neurons/physiology , Action Potentials/physiology , Animals , Arginine Vasopressin/genetics , Arginine Vasopressin/metabolism , Brain Mapping , Cell Plasticity/physiology , Circadian Rhythm/physiology , Female , Kisspeptins/metabolism , Mice , Mice, Transgenic , Neurons/metabolism , Proestrus/physiology , Suprachiasmatic Nucleus/metabolism , Suprachiasmatic Nucleus/physiology
5.
Psychopharmacology (Berl) ; 238(4): 969-978, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33404736

ABSTRACT

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.


Subject(s)
Estradiol/administration & dosage , Heroin/administration & dosage , Progesterone/administration & dosage , Animals , Estradiol/pharmacology , Estrus/physiology , Female , Heroin/pharmacology , Mifepristone/administration & dosage , Mifepristone/pharmacology , Proestrus/physiology , Progesterone/pharmacology , Rats , Rats, Long-Evans
6.
Croat Med J ; 61(4): 346-353, 2020 Aug 31.
Article in English | MEDLINE | ID: mdl-32881433

ABSTRACT

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.


Subject(s)
Adipose Tissue, Brown/metabolism , Estrous Cycle/physiology , Skin Temperature/physiology , Thermography/methods , Animals , Animals, Laboratory , Estrus/physiology , Female , Infrared Rays , Mice , Mice, Inbred C57BL , Proestrus/physiology
7.
J Am Assoc Lab Anim Sci ; 59(3): 282-287, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32241334

ABSTRACT

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.


Subject(s)
Estrous Cycle , Rats/physiology , Reproductive Techniques/veterinary , Vagina/physiology , Animal Technicians , Animals , Electric Impedance , Female , Luteinizing Hormone/agonists , Male , Pregnancy , Proestrus/physiology
8.
Sci Rep ; 9(1): 18403, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31804540

ABSTRACT

The temporomandibular muscle dysfunction is characterized by myofascial pain and is more prevalent in women of reproductive age. Sex steroid hormones are hypothetically involved in the dysfunction, but few are the studies of steroid receptors in masticatory and mastication-related muscles. Our aim was to determine estrogen and testosterone receptor expression in rat masticatory and mastication-related muscles within the context of age and gender. Twelve rats were equally divided into four groups: (a) 10-month-old females; (b) 10-month-old males; (c) 24-month-old females; and (d) 24-month-old males. Euthanasia of the females was performed in the proestrous phase (vaginal smears) and the masticatory and accessory muscles were removed for immunohistochemical analysis. Statistical analysis was performed with ANOVA and the Tukey test. Estrogen receptor expression was similarly low in all muscles and groups. Testosterone receptor expression in the Masseter muscle of the 24-month-old male rats was higher than that in the other groups and significantly superior to its expression in the Posterior Digastric muscle. In short, testosterone receptor expression was highest in old male rats. If we generalize to humans, this fact could indicate age- and sex-related hormonal influence on temporomandibular muscle dysfunction. Further studies, however, are necessary to strengthen this hypothesis.


Subject(s)
Aging/genetics , Masticatory Muscles/metabolism , Receptors, Androgen/genetics , Receptors, Estrogen/genetics , Animals , Estrogens/blood , Female , Gene Expression Regulation, Developmental , Male , Mastication/physiology , Masticatory Muscles/growth & development , Proestrus/physiology , Rats , Rats, Wistar , Receptors, Androgen/metabolism , Receptors, Estrogen/metabolism , Signal Transduction , Testosterone/blood
9.
Respir Physiol Neurobiol ; 264: 1-7, 2019 06.
Article in English | MEDLINE | ID: mdl-30898577

ABSTRACT

Ventilatory long-term facilitation (vLTF) is a form of respiratory plasticity characterized by a progressive and sustained increase in minute ventilation over time following acute, intermittent hypoxia (AIH). Though vLTF has been repeatedly demonstrated in adult males (rats and humans), few studies have assessed vLTF in adult females and no studies have explored differential expression of vLTF across the normal female estrous cycle. We recently reported that AIH-induced plasticity of phrenic motor output (phrenic long-term facilitation, pLTF), a phenotypically similar form of respiratory plasticity presenting as a sustained increase in phrenic nerve amplitude, develops in adult female rats only during the proestrus stage of the estrous cycle, notable for high levels of serum estrogen. Here, we tested the hypothesis that AIH-induced vLTF would also be estrous-stage dependent; developing in female rats during proestrus, but not estrus. Barometric plethysmography in adult (4-5 months), normally cycling female rats revealed a progressive increase in minute ventilation for 60 min following AIH (5 × 5 min episodes; 10% O2) during proestrus indicative of vLTF, while estrus rats showed no changes in minute ventilation over the same time period. The development of vLTF in proestrus rats was driven by changes in tidal volume production versus respiratory frequency consistent with prior studies. These data are the first to investigate differential vLTF expression across the estrous cycle in adult female rats and highlight the importance of female estrous cycle stage as a critical physiological variable to consider in studies of AIH-induced plasticity.


Subject(s)
Estrous Cycle/physiology , Hypoxia/physiopathology , Respiratory Rate/physiology , Animals , Disease Models, Animal , Female , Plethysmography , Proestrus/physiology , Rats , Rats, Sprague-Dawley , Tidal Volume/physiology
10.
Pesqui. vet. bras ; 38(8): 1518-1527, Aug. 2018. tab, graf
Article in Portuguese | LILACS, VETINDEX | ID: biblio-976480

ABSTRACT

Objetivou-se avaliar o efeito de uma ou duas doses de prostaglandina F2α (PGF2α) associada ou não a gonadotrofina coriônica equina (eCG) sobre a dinâmica folicular, a função luteal pré-ovulatória, assim como as características morfofuncionais pós-ovulatórias do corpo lúteo (CL) em fêmeas mestiças cíclicas submetidas a um protocolo de inseminação artificial em tempo fixo (IATF). Para tanto, 29 vacas 3/4 Gir x Holandês multíparas foram submetidas ao exame de ultrassonografia (US) transretal e após a detecção do CL iniciou-se um protocolo de IATF em um dia denominado zero (D0), por meio da inserção do implante de progesterona (P4) associado à aplicação de 2,0mg de benzoato de estradiol. No D7 esses animais receberam 12,5mg de dinoprost trometamina. No D9 realizou a remoção dos dispositivos de P4 e aplicou 0,6mg de cipionato de estradiol. Nesse momento, as fêmeas foram subdivididas nos seguintes tratamentos: Grupo Controle (n=7), foi administrado 2,5mL de solução fisiológica; Grupo 2PGF (n=7), aplicou 12,5mg de dinoprost trometamina; Grupo eCG (n=7), administrou-se 300UI de eCG; Grupo 2PGF+eCG (n=8), realizou a aplicação de 300UI de eCG e 12,5mg de dinoprost trometamina. Para avaliar a dinâmica folicular foram realizados exames de US em modo B e power doppler (Mindray Z5, Shenzhen, China) a cada 12h do D7 até o momento da ovulação ou 96h após a remoção dos implantes de P4, mensurando-se o diâmetro folicular (DFOL), a área da parede folicular (AFOL) e a área de perfusão sanguínea da parede folicular (VFOL). Concomitante a cada exame, foram coletadas amostras de sangue sendo determinada a concentração sérica de P4 pré-ovulatória por meio da metodologia de quimioluminescência. No D24 foi realizada a US modo B e doppler analisando-se o diâmetro luteal (DCL), área luteal (ACL) e área de perfusão sanguínea do CL (VCL), assim como, foi coletada amostra de sangue para averiguar a concentração sérica de P4 pós-ovulatória. Os dados foram avaliados pelo Two-way ANOVA e análise de medidas repetidas considerando os efeitos do eCG, 2PGF e interação eCG*2PGF, P<0,05. Não houve diferença significativa entre os protocolos de sincronização para as variáveis DFOL, AFOL e VFOL ao longo do tempo da dinâmica folicular. Os grupos experimentais apresentaram uma concentração sérica de P4 pré-ovulatória semelhante em cada momento da avaliação. Não foi observada distinção da ACL e VCL entre os tratamentos hormonais, contudo o Grupo eCG demonstrou tendência (P=0,08) a apresentar maior DCL em relação ao Grupo 2PGF e 2PGF+eCG. Adicionalmente a estes achados, também foi constatado tendência (P=0,07) a maiores concentrações de progesterona no dia 24 do protocolo nos animais do Grupo eCG (11,00±3,32ng/mL) em relação ao Grupo 2PGF (6,37±1,31ng/mL), enquanto o Controle e 2PGF+eCG demonstraram resultados intermediários que se assemelham a ambos os grupos, com concentrações de 8,43±3,85 e 9,18±2,82ng/mL, respectivamente. As tentativas de ajustes no proestro foram incapazes de melhorar a qualidade folicular e minimizar a função luteal pré-ovulatória, assim como não incrementaram a morfologia do CL e a função luteal pós-ovulatória, sugerindo que em animais cíclicos mestiços protocolos de IATF com a utilização de uma única dose PGF2α e sem o suporte gonadotrófico da eCG parece promover adequada resposta folicular e luteal.(AU)


The study aimed to evaluate the effect of one or two prostaglandin doses F2α (PGF2a) with or without equine chorionic gonadotropin (eCG) in the follicular dynamics, the preovulatory luteal function, as well as the structural and functional characteristics post-ovulatory of the corpus luteum (CL) in cyclic crossbred females subjected to a fixed time artificial insemination (FTAI) protocol. For this, 29 multiparous 3/4 Gyr x Holstein cows were subjected to transrectal ultrasound examination (US) and upon detection of CL initiated a FTAI protocol on day called zero (D0) by the insertion of progesterone implant (P4) associated with the application of 2.0mg estradiol benzoate. On D7, these animals received 12.5mg of dinoprost tromethamine. At D9 happened the removal of the P4 devices and was applied 0.6mg of estradiol cypionate. At that time, the females were divided into the following treatments: control group (n=7) - which received 2.5mL of saline solution, 2PGF group (n=7) - received 12.5mg of dinoprost tromethamine, eCG group (n=7) - was administered 300IU eCG and eCG+2PGF group (n=8) - which received 300 IU eCG and 12.5mg of dinoprost tromethamine. To assess follicular dynamics were performed US scans B-mode and power doppler (Mindray Z5, Shenzhen, China) each 12h on D7 until the time of ovulation or until 96h after removal of the P4 implants, considering the follicular diameter (DFOL), the area of the follicular wall (AFOL) and the blood perfusion area of the follicular wall (VFOL). Concomitant with each test, blood samples were collected to determine the serum concentration of P4 preovulatory by chemiluminescence methodology. In D24 had held US B-mode and doppler to analyse the luteal diameter (DCL), luteal area (ACL) and blood perfusion area CL (VCL). Also, a blood sample was collected to determine the serum concentration of P4 post-ovulatory. All data was evaluated by Two-way ANOVA and repeated measures analysis considering the effects of eCG, 2PGF and eCG*2PGF, P<0.05. There was not significant difference between the synchronization protocols for DFOL, AFOL and VFOL variables over time of follicular dynamics. Experimental groups had a serum concentration of P4 preovulatory similar in every moment of evaluation. There wasn't distinction of ACL and VCL between hormone treatments. However, the eCG group showed a tendency (P=0.08) to present higher DCL compared to the 2PGF and 2PGF+eCG groups. In addition to these findings, there was also a tendency (P=0.07) to higher concentrations of P4 on D24 of the protocol in the animals of the eCG group (11.00±3.32ng/mL) compared to the 2PGF group (6,37±1.31ng/mL), meanwhile the Control and 2PGF+eCG showed intermediate results that resembled both groups, with concentrations of 8.43±3.85 and 9.18±2.82ng/mL, respectively. Attempts to adjust proestrus were unable to improve follicular quality and minimize preovulatory luteal function, nor did they increase CL morphology and post-ovulatory luteal function, suggesting that in cyclic animals, FTAI protocols using a single PGF2α dose and without the gonadotrophic support of eCG seems to promote adequate follicular and luteal responses.(AU)


Subject(s)
Animals , Proestrus/physiology , Cattle/metabolism , Gonadotropins, Equine/analysis , Estrus Synchronization
11.
Reprod Fertil Dev ; 30(11): 1541-1552, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29778102

ABSTRACT

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.


Subject(s)
Estradiol/analogs & derivatives , Estrus Synchronization/physiology , Ovary/physiology , Proestrus/physiology , Progesterone/administration & dosage , Uterus/physiology , Animals , Cattle , Estradiol/administration & dosage , Estradiol/blood , Estrus Synchronization/drug effects , Female , Ovarian Follicle/diagnostic imaging , Ovarian Follicle/drug effects , Ovary/diagnostic imaging , Ovary/drug effects , Pregnancy , Proestrus/drug effects , Progesterone/blood , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Uterus/diagnostic imaging , Uterus/drug effects
12.
Behav Brain Res ; 339: 124-129, 2018 Feb 26.
Article in English | MEDLINE | ID: mdl-29180134

ABSTRACT

Females are an under-represented research model and the mechanisms through which sleep loss impairs cognition are not clear. Since levels of reproductive hormones and the estrous cycle are sensitive to sleep loss and necessary for learning and memory, we hypothesized that sleep deprivation impacts learning and memory in female mice by interfering with the estrous cycle. We used the object recognition task to assess learning and memory in female mice during separate phases of the estrous cycle and after sleep loss. Mice in metestrus/diestrus attended to sample objects less than mice in proestrus/estrus during object acquisition, the first phase of the object recognition task. Subsequently, during the recognition phase of the task, only mice in proestrus/estrus displayed a preference for the novel object. Sleep deprivation for 12h immediately before the object recognition task reduced time attending to sample objects and novel object preference for mice in proestrus/estrus, without changing length of the estrous cycle. These results show that sleep deprived mice in proestrus/estrus had learning deficits and memory impairments, like mice in metestrus/diestrus. Since sleep deprivation did not disrupt the estrous cycle, however, results did not support the hypothesis. Cognitive impairments due to acute sleep loss were not due to alterations to the estrous cycle.


Subject(s)
Estrous Cycle/physiology , Learning/physiology , Memory/physiology , Metestrus/physiology , Sleep Deprivation/physiopathology , Animals , Behavior, Animal/physiology , Female , Mice, Inbred C57BL , Proestrus/physiology
13.
J Neurosci ; 38(5): 1061-1072, 2018 01 31.
Article in English | MEDLINE | ID: mdl-29114074

ABSTRACT

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.


Subject(s)
Estradiol/pharmacology , Glutamates/physiology , Hypothalamus/cytology , Hypothalamus/physiology , Kisspeptins/physiology , Neurons/physiology , Receptors, Estrogen/drug effects , Synaptic Transmission/physiology , Animals , Arcuate Nucleus of Hypothalamus/physiology , Dynorphins/pharmacology , Female , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Hypothalamus/drug effects , Luteinizing Hormone/physiology , Mice , Midline Thalamic Nuclei/physiology , Neurons/drug effects , Pituitary Gland/drug effects , Pituitary Gland/physiology , Proestrus/physiology , Receptors, Ionotropic Glutamate/drug effects , Receptors, Ionotropic Glutamate/physiology , Synaptic Transmission/drug effects , ERRalpha Estrogen-Related Receptor
14.
J Cell Physiol ; 233(9): 6591-6603, 2018 09.
Article in English | MEDLINE | ID: mdl-29115663

ABSTRACT

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.


Subject(s)
Annexin A1/metabolism , Embryo Implantation/physiology , Animals , Estrous Cycle/metabolism , Estrous Cycle/physiology , Female , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Models, Animal , Pregnancy , Proestrus/metabolism , Proestrus/physiology , Sex Ratio , Uterus/metabolism , Uterus/physiology , X Chromosome/metabolism , X Chromosome/physiology , Y Chromosome/metabolism , Y Chromosome/physiology
15.
Niger J Physiol Sci ; 32(1): 13-20, 2017 Jun 30.
Article in English | MEDLINE | ID: mdl-29134972

ABSTRACT

Stress has been acknowledged as one of the aetiologies of female reproductive dysfunction, yet the mechanismsinvolved are not totally elucidated. Based on the paucity of information on how predator-induced stress (PS) affects oestrouscycle in rats, this study was designed to investigate the effect of PS on the oestrous cycle in rats. Forty-eight (48) SpragueDawley rats were used for this study. They were randomly divided into Control and PS group. Each group was divided intofour subgroups (n=6/group) according to the phases of oestrous cycle. Stress was induced by exposing rats to cat (predator)for 60 minutes/day for 14 consecutive days. PS caused significant disruption of the oestrous cycle. In animals subjected toPS at proestrus (PS-proestrus) and oestrus (PS-oestrus), percentage occurrence of proestrus, oestrus and metestrus phaseswere significantly reduced compared with control. In animals subjected to PS at metestrus (PS-metestrus) and diestrus (PSdiestrus), percentage occurrence of oestrus phase was not significantly affected. In all animals exposed to PS, percentageoccurrence of diestrus was significantly increased regardless of the phase of first exposure compared with control.Corticosterone and prolactin levels were significantly elevated in PS groups compared with control. Progesterone wassignificantly increased in animals at diestrus phase compared with oestrus phase and respective phases in control. Oestradiolwas significantly reduced in PS group compared with control at oestrus phase but not significantly different at diestrus phase.Luteinizing hormone (LH) and follicle stimulating hormone (FSH) levels were significantly lower in PS groups at oestrusphase compared with diestrus phase. This study shows that PS disrupts the oestrous cycle secondary to perturbation ofhormonal control of female reproduction and is influenced by the phase at first exposure to stress.


Subject(s)
Estrous Cycle/physiology , Estrus/physiology , Luteinizing Hormone/metabolism , Stress, Physiological/physiology , Animals , Cats , Estradiol/metabolism , Female , Follicle Stimulating Hormone/metabolism , Proestrus/physiology , Progesterone/metabolism , Rats
16.
Endocrinology ; 158(10): 3249-3258, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28977586

ABSTRACT

Sex differences exist in chronic pain pathologies, and gonadal estradiol (E2) alters the pain sensation. The nocisensor transient receptor potential vanilloid 1 (TRPV1) receptor plays a critical role in triggering pain. Here we examined the impact of E2 on the function of TRPV1 receptor in mice sensory neurons in vitro and in vivo. Both mechano- and thermonociceptive thresholds of the plantar surface of the paw of female mice were significantly lower in proestrus compared with the estrus phase. These thresholds were higher in ovariectomized (OVX) mice and significantly lower in sham-operated mice in proestrus compared with the sham-operated mice in estrus phase. This difference was absent in TRPV1 receptor-deficient mice. Furthermore, E2 potentiated the TRPV1 receptor activation-induced mechanical hyperalgesia in OVX mice. Long pretreatment (14 hours) with E2 induced a significant increase in TRPV1 receptor messenger RNA expression and abolished the capsaicin-induced TRPV1 receptor desensitization in primary sensory neurons. The short E2 incubation (10 minutes) also prevented the desensitization, which reverted after coadministration of E2 and the tropomyosin-related kinase A (TrkA) receptor inhibitor. Our study provides in vivo and in vitro evidence for E2-induced TRPV1 receptor upregulation and sensitization mediated by TrkAR via E2-induced genomic and nongenomic mechanisms. The sensitization and upregulation of TRPV1 receptor by E2 in sensory neurons may explain the greater pain sensitivity in female mice.


Subject(s)
Estradiol/pharmacology , Pain/physiopathology , TRPV Cation Channels/drug effects , TRPV Cation Channels/physiology , Animals , Capsaicin/pharmacology , Cells, Cultured , Drug Tolerance , Estrus/physiology , Female , Gene Expression/drug effects , Hot Temperature , Male , Mechanoreceptors/drug effects , Mechanoreceptors/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nociceptors/drug effects , Nociceptors/physiology , Ovariectomy , Proestrus/physiology , RNA, Messenger/analysis , Sensory Receptor Cells/chemistry , Sensory Receptor Cells/physiology , Sex Characteristics , TRPV Cation Channels/genetics , Up-Regulation/drug effects
17.
Microsc Res Tech ; 80(2): 167-176, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27717109

ABSTRACT

The current study aimed to determine if characteristics observed in vaginal cytology during the estrous cycle of female SYT cavies corresponded with proliferation of the vaginal epithelium, characterized by proliferating cell nuclear antigen (PCNA) immunolocalization, and with follicular development at different phases of the estrous cycle. After determining estrous cycle phases by vaginal cytology, females were euthanized at metestrus, diestrus, proestrus, and estrus. Histological study of the vaginal epithelium and ovary were then performed. Immunohistochemistry for PCNA in vaginal tissue at each cycle phase was also performed. Superficial cornified cells and early post-ovulatory follicles were found at estrus. Few nuclei below the enucleate superficial cells were immunoreactive to PCNA. At metestrus, the vaginal epithelium underwent desquamation and lost the superficial cornified cells; basal and intermediate cells appeared, and the post-ovulatory follicle formed an early corpus luteum. No PCNA immunoreactivity was observed. At diestrus, the corpus luteum was developed, and the vaginal epithelium contained basal and intermediate cells. There was PCNA immunoreactivity in the cellular nucleus in the germinative stratum of the epithelium. Because of the growth and maturation of ovarian follicles, the vaginal epithelium suffered intense proliferation at proestrus. Vaginal cytology revealed large intermediate cells and nucleated and enucleated superficial cornified cells. In the ovary, mature follicles were present. More apparent immunoreactivity of PCNA in the germinative layer was found. In summary, we inferred that vaginal exfoliative findings matched the proliferation process of the vaginal epithelium. PCNA immunolocalization occurred as well as corresponding follicular development in the ovaries.


Subject(s)
Estrous Cycle/physiology , Ovarian Follicle/cytology , Ovarian Follicle/physiology , Rodentia/physiology , Vagina/cytology , Animals , Cell Proliferation , Diestrus/physiology , Epithelium/immunology , Epithelium/physiology , Estrus/physiology , Female , Histological Techniques , Immunohistochemistry , Ovarian Follicle/ultrastructure , Ovary/cytology , Ovary/ultrastructure , Proestrus/physiology , Proliferating Cell Nuclear Antigen/analysis , Proliferating Cell Nuclear Antigen/immunology , Rodentia/anatomy & histology , Vagina/physiology
18.
Endocrinology ; 158(2): 356-366, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27911605

ABSTRACT

During the female reproductive cycle, estradiol exerts negative and positive feedback at both the central level to alter gonadotropin-releasing hormone (GnRH) release and at the pituitary to affect response to GnRH. Many studies of the neurobiologic mechanisms underlying estradiol feedback have been done on ovariectomized, estradiol-replaced (OVX+E) mice. In this model, GnRH neuron activity depends on estradiol and time of day, increasing in estradiol-treated mice in the late afternoon, coincident with a daily luteinizing hormone (LH) surge. Amplitude of this surge appears lower than in proestrous mice, perhaps because other ovarian factors are not replaced. We hypothesized GnRH neuron activity is greater during the proestrous-preovulatory surge than the estradiol-induced surge. GnRH neuron activity was monitored by extracellular recordings from fluorescently tagged GnRH neurons in brain slices in the late afternoon from diestrous, proestrous, and OVX+E mice. Mean GnRH neuron firing rate was low on diestrus; firing rate was similarly increased in proestrous and OVX+E mice. Bursts of action potentials have been associated with hormone release in neuroendocrine systems. Examination of the patterning of action potentials revealed a shift toward longer burst duration in proestrous mice, whereas intervals between spikes were shorter in OVX+E mice. LH response to an early afternoon injection of GnRH was greater in proestrous than diestrous or OVX+E mice. These observations suggest the lower LH surge amplitude observed in the OVX+E model is likely not attributable to altered mean GnRH neuron activity, but because of reduced pituitary sensitivity, subtle shifts in action potential pattern, and/or excitation-secretion coupling in GnRH neurons.


Subject(s)
Estradiol/physiology , Gonadotropin-Releasing Hormone/physiology , Luteinizing Hormone/blood , Pituitary Gland/physiology , Proestrus/physiology , Action Potentials , Animals , Female , Mice, Inbred C57BL , Neurons/metabolism
19.
Reprod Sci ; 24(6): 844-855, 2017 06.
Article in English | MEDLINE | ID: mdl-27688242

ABSTRACT

The aims of the present study were to analyze if the superior ovarian nerve (SON) plays a role in the neural signals from suprachiasmatic nucleus (SCN) that lead to ovulation and ovarian steroids secretion on proestrus day. Rats on proestrus day were treated at 11.00 to 11.30 or 17.00 to 17.30 hours with 1 of the 3 experimental procedures (1) unilateral or bilateral SON sectioning, (2) unilateral or bilateral injury to the SCN, or (3) unilateral injury to the SCN followed by unilateral sectioning of the SON ipsilateral to the treated SCN. Treatments were evaluated 24 hours after surgical procedures. Compared to laparotomized animals, right or bilateral SON sectioning treatment at 17.00 hours resulted in lower ovulation rates and number of ova shed by the right ovary. The ovaries of nonovulating animals showed early follicular luteinization signs and trapped ova. Bilateral SCN injury treatment at 11.00 hours resulted in anovulation; whereas right SCN injury treatment, with or without right SON sectioning, resulted in a lower number of ova shed. Injecting luteinizing hormone-releasing hormone to animals with bilateral SCN injury restored ovulation. In rats with unilateral or bilateral SON sectioning, or with injury to the SCN with or without unilateral sectioning of the SON, the effects on hormone levels depended of the hormone studied and the time of day treatment was performed. The present results suggest that on proestrus day, the role of the right or both SON in ovulation and steroid hormone secretion regulation takes place through different neuroendocrine mechanisms from SCN.


Subject(s)
Estradiol/blood , Ovary/innervation , Ovary/pathology , Ovulation/physiology , Proestrus/physiology , Progesterone/blood , Suprachiasmatic Nucleus/physiology , Animals , Female , Gonadotropin-Releasing Hormone/pharmacology , Ovary/drug effects , Ovulation/drug effects , Proestrus/drug effects , Rats , Testosterone/blood
20.
Neuroscience ; 343: 384-397, 2017 02 20.
Article in English | MEDLINE | ID: mdl-28012870

ABSTRACT

Stress and withdrawal of female reproductive hormones are known risk factors of postpartum depression. Although both of these factors are capable of powerfully modulating neuronal plasticity, there is no direct electron microscopic evidence of hippocampal spine synapse remodeling in postpartum depression. To address this issue, hormonal conditions of pregnancy and postpartum period were simulated in ovariectomized adult female Sprague-Dawley rats (n=76). The number of hippocampal spine synapses and the depressive behavior of rats in an active escape task were investigated in untreated control, hormone-withdrawn 'postpartum', simulated proestrus, and hormone-treated 'postpartum' animals. After 'postpartum' withdrawal of gonadal steroids, inescapable stress caused a loss of hippocampal spine synapses, which was related to poor escape performance in hormone-withdrawn 'postpartum' females. These responses were equivalent with the changes observed in untreated controls that is an established animal model of major depression. Maintaining proestrus levels of ovarian hormones during 'postpartum' stress exposure did not affect synaptic and behavioral responses to inescapable stress in simulated proestrus animals. By contrast, maintaining pregnancy levels of estradiol and progesterone during 'postpartum' stress exposure completely prevented the stress-induced loss of hippocampal spine synapses, which was associated with improved escape performance in hormone-treated 'postpartum' females. This protective effect appears to be mediated by a muted stress response as measured by serum corticosterone concentrations. In line with our emerging 'synaptogenic hypothesis' of depression, the loss of hippocampal spine synapses may be a novel perspective both in the pathomechanism and in the clinical management of postpartum affective illness.


Subject(s)
Depression, Postpartum/pathology , Depressive Disorder, Major/pathology , Hippocampus/pathology , Neuronal Plasticity , Synapses/pathology , Animals , Corticosterone/blood , Depression, Postpartum/metabolism , Depressive Disorder, Major/metabolism , Disease Models, Animal , Estradiol/administration & dosage , Estradiol/metabolism , Female , Hippocampus/metabolism , Neuronal Plasticity/physiology , Ovariectomy , Postpartum Period , Proestrus/physiology , Progesterone/administration & dosage , Progesterone/metabolism , Rats, Sprague-Dawley , Synapses/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...