Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.109
Filtrar
1.
Theriogenology ; 219: 157-166, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38432143

RESUMO

To understand better the role that kisspeptin plays in regulating seasonal and estrous cycle changes in the mare, this study investigated the number, location and interactions between GnRH, kisspeptin and RFRP-3 neurons in the equine hypothalamus. Hypothalami were collected from mares during the non-breeding season, vernal transition and various stages of the breeding season. Fluorescent immunohistochemistry was used to label the neuropeptides of interest. GnRH cells were observed primarily in the arcuate nucleus (ARC), while very few labeled cells were identified in the pre-optic area (POA). Kisspeptin cells were identified primarily in the ARC, with a small number of cells observed dorsal to the ARC, surrounding the third ventricle (3V). The mean number of kisspeptin cells varied between animals and typically showed no pattern associated with season or stage of estrous cycle, but a seasonal difference was identified in the ARC population. Small numbers of RFRP-3 cells were observed in the ARC, ventromedial hypothalamus (VMH) and dorsomedial hypothalamus (DMH). The mean number of RFRP-3 cells appeared higher in pre-ovulatory animals compared to all other stages. The percentage of GnRH cell bodies with kisspeptin appositions did not change with season or stage of estrous cycle. The percentage of kisspeptin cells receiving inputs from RFRP-3 fibers did not vary with season or stage of estrous cycle. These interactions suggest the possibility of the presence of an ultra-short loop feedback system between these three peptides. The changes in RFRP-3 neurons suggest the possibility of a role in the regulation of reproduction in the horse, but it is unlikely to be as a gonadotropin inhibitory factor.


Assuntos
Hormônio Liberador de Gonadotropina , Neuropeptídeos , Cavalos , Animais , Feminino , Hormônio Liberador de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Estações do Ano , Neuropeptídeos/fisiologia , Hipotálamo/metabolismo , Ciclo Estral/fisiologia , Neurônios
2.
J Equine Vet Sci ; 135: 105034, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38428754

RESUMO

Gut microbiota plays a crucial role in various physiological processes, including the regulation of the reproductive system and steroid sex hormones. Throughout the normal estrous cycle of healthy mares, the levels of estradiol-17ß (E2) and progesterone (P4) in the blood exhibit periodic changes. To investigate the relationship between cyclic changes in steroid sex hormones and the gut microbiome of mares, we analyzed the fecal microbiota composition in healthy mares during the typical estrous cycle. Blood and fecal samples from five healthy mares were collected, E2 and P4 levels in serum were analyzed using radioimmunoassay (RIA), and the gut microbiome was analyzed by 16S rRNA sequencing. The overall richness and composition of the gut microbiota remained relatively stable during the normal estrous cycle in mares. The Linear Discriminant Analysis Effect Size analysis of the microbial composition during the follicular and luteal phases identified the Rhodococcus genus as differentially abundant. These findings indicate that the mare's gut microbiota's significant composition remains consistent throughout the estrous cycle. At the same time, specific low-abundance pathogenic bacteria exhibit changes that align with sexual hormonal fluctuations.


Assuntos
Ciclo Estral , Microbiota , Cavalos , Animais , Feminino , RNA Ribossômico 16S/genética , Ciclo Estral/fisiologia , Progesterona , Hormônios Esteroides Gonadais
3.
Behav Brain Res ; 461: 114860, 2024 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-38216058

RESUMO

Despite known sex differences in brain function, female subjects are underrepresented in preclinical neuroscience research. This is driven in part by concerns about variability arising from estrous cycle-related hormone fluctuations, especially in fear- and anxiety-related research where there are conflicting reports as to whether and how the cycle influences behavior. The inconsistency may arise from a lack of common standards for tracking and reporting the cycle as opposed to inherent unpredictability in the cycle itself. The rat estrous cycle is conventionally tracked by assigning vaginal cytology smears to one of four qualitatively-defined stages. Although the cytology stages are of unequal length, the stage names are often, but not always, used to refer to the four cycle days. Subjective staging criteria and inconsistent use of terminology are not necessarily a problem in research on the cycle itself, but can lead to irreproducibility in neuroscience studies that treat the stages as independent grouping factors. We propose the explicit use of cycle days as independent variables, which we term Track-by-Day to differentiate it from traditional stage-based tracking, and that days be indexed to the only cytology feature that is a direct and rapid consequence of a hormonal event: a cornified cell layer formed in response to the pre-ovulatory 17ß-estradiol peak. Here we demonstrate that cycle length is robustly regular with this method, and that the method outperforms traditional staging in detecting estrous cycle effects on Pavlovian fear conditioning and on a separate proxy for hormonal changes, uterine histology.


Assuntos
Ciclo Estral , Vagina , Humanos , Ratos , Feminino , Masculino , Animais , Ciclo Estral/fisiologia , Vagina/fisiologia , Estradiol/farmacologia , Medo/fisiologia
4.
Endocrinology ; 165(3)2024 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-38279940

RESUMO

The arcuate nucleus kisspeptin (ARNKISS) neurons represent the GnRH pulse generator that likely drives pulsatile gonadotropin secretion in all mammals. Using an improved GCaMP fiber photometry system enabling long-term continuous recordings, we aimed to establish a definitive profile of ARNKISS neuronal activity across the murine estrous cycle. As noted previously, a substantial reduction in the frequency of ARNKISS neuron synchronization events (SEs) occurs on late proestrus and extends into estrus. The SE amplitude remains constant throughout the cycle. During metestrus, we unexpectedly detected many multipeak SEs where many SEs occurred rapidly, within 160 seconds of each other. By applying a machine learning-based, k-means clustering analysis, we were further able to detect substantial within-stage variability in the patterns of pulse generator activity. Estrous cycle-dependent changes in SE activity occurred around the time of lights on and off. We also find that a mild stressor such as vaginal lavage reduces ARNKISS neuron SE frequency for up to 3 hours. These observations provide a comprehensive account of ARNKISS neuron activity across the estrous cycle, highlight a new pattern of multipeak SE activity, and introduce a new k-means clustering approach for analyzing ARNKISS neuron population behavior.


Assuntos
Hormônio Liberador de Gonadotropina , Hormônio Luteinizante , Animais , Feminino , Camundongos , Núcleo Arqueado do Hipotálamo/metabolismo , Ciclo Estral/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Neurônios/metabolismo
5.
Physiol Genomics ; 56(1): 74-97, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-37694291

RESUMO

Conserved in female reproduction across all mammalian species is the estrous cycle and its regulation by the hypothalamic-pituitary-gonadal (HPG) axis, a collective of intersected hormonal events that are crucial for ensuring uterine fertility. Nonetheless, knowledge of the direct mediators that synchronously shape the uterine microenvironment for successive yet distinct events, such as the transit of sperm and support for progressive stages of preimplantation embryo development, remain principally deficient. Toward understanding the timed endometrial outputs that permit luminal events as directed by the estrous cycle, we used Bovidae as a model system to uniquely surface sample and study temporal shifts to in vivo endometrial transcripts that encode for proteins destined to be secreted. The results revealed the full quantitative profile of endometrial components that shape the uterine luminal microenvironment at distinct phases of the estrous cycle (estrus, metestrus, diestrus, and proestrus). In interpreting this comprehensive log of stage-specific endometrial secretions, we define the "uterine secretory cycle" and extract a predictive understanding of recurring physiological actions regulated within the uterine lumen in anticipation of sperm and preimplantation embryonic stages. This repetitive microenvironmental preparedness to sequentially provide operative support was a stable intrinsic framework, with only limited responses to sperm or embryos if encountered in the lumen within the cyclic time period. In uncovering the secretory cycle and unraveling realistic biological processes, we present novel foundational knowledge of terminal effectors controlled by the HPG axis to direct a recurring sequence of vital functions within the uterine lumen.NEW & NOTEWORTHY This study unravels the recurring sequence of changes within the uterus that supports vital functions (sperm transit and development of preimplantation embryonic stages) during the reproductive cycle in female Ruminantia. These data present new systems knowledge in uterine reproductive physiology crucial for setting up in vitro biomimicry and artificial environments for assisted reproduction technologies for a range of mammalian species.


Assuntos
Sêmen , Útero , Gravidez , Animais , Feminino , Masculino , Útero/metabolismo , Endométrio , Ciclo Estral/fisiologia , Estro , Mamíferos
6.
Elife ; 122023 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-38126277

RESUMO

Evidence suggests that estradiol-sensing preoptic area GABA neurons are involved in the preovulatory surge mechanism necessary for ovulation. In vivo CRISPR-Cas9 editing was used to achieve a 60-70% knockdown in estrogen receptor alpha (ESR1) expression by GABA neurons located within the regions of the rostral periventricular area of the third ventricle (RP3V) and medial preoptic nuclei (MPN) in adult female mice. Mice exhibited variable reproductive phenotypes with the only significant finding being mice with bilateral ESR1 deletion in RP3V GABA neurons having reduced cFos expression in gonadotropin-releasing hormone (GnRH) neurons at the time of the surge. One sub-population of RP3V GABA neurons expresses kisspeptin. Re-grouping ESR1-edited mice on the basis of their RP3V kisspeptin expression revealed a highly consistent phenotype; mice with a near-complete loss of kisspeptin immunoreactivity displayed constant estrus and failed to exhibit surge activation but retained pulsatile luteinizing hormone (LH) secretion. These observations demonstrate that ESR1-expressing GABA-kisspeptin neurons in the RP3V are essential for the murine preovulatory LH surge mechanism.


Assuntos
Sistemas CRISPR-Cas , Kisspeptinas , Camundongos , Feminino , Animais , Kisspeptinas/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios GABAérgicos/metabolismo , Ciclo Estral/fisiologia , Ácido gama-Aminobutírico/metabolismo
7.
Sci Rep ; 13(1): 22353, 2023 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-38102308

RESUMO

Dynamic functional changes in the oviductal microenvironment are the prerequisite for the establishment of pregnancy. The objective of this study was to gain the first insights into oestrous cycle-dependent dynamics of polymorph nuclear neutrophils (PMN) and the mRNA abundance of selected genes and their correlations in the oviduct of living cows. Mini-cytobrush samples were taken from the oviducts of healthy heifers (n = 6) and cows (n = 7) during the follicular (FOL) and luteal phase (LUT) by transvaginal endoscopy. Total RNA was isolated from the samples and subjected to reverse transcription-quantitative PCR for selected pro-inflammatory factors, glycoproteins, and a metabolic marker. The percentage of PMN was determined by cytological examination. The mean PMN percentage was 2.8-fold greater during LUT than FOL. During LUT, significantly greater mRNA abundance of the pro-inflammatory factors IL1B, CXCL1, CXCL3, and CXCL8 was observed. The OVGP1 mRNA abundance was twice as high during FOL than in LUT. Pearson correlation, principal component analysis and heatmap analyses indicated characteristic functional patterns with strong correlations among investigated factors. Using this novel approach, we illustrate complex physiological dynamics and interactions of the mRNA expression of pro-inflammatory factors, mucins, OVGP1, and PMN in the oviduct during the oestrous cycle.


Assuntos
Mucinas , Neutrófilos , Gravidez , Humanos , Bovinos , Animais , Feminino , Mucinas/genética , Mucinas/metabolismo , Neutrófilos/metabolismo , Fase Luteal , Ciclo Estral/fisiologia , Tubas Uterinas/metabolismo , Oviductos/metabolismo , RNA Mensageiro/metabolismo , Glicoproteínas/metabolismo
8.
Endocrinology ; 165(1)2023 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-37967240

RESUMO

Serum sex steroid levels fluctuate throughout the reproductive cycle. However, the degree to which sex steroid tissue content mimics circulating content is unknown. Understanding the flux and physiological quantity of tissue steroid content is imperative for targeted hormonal therapy development. Utilizing a gold-standard ultrasensitive liquid chromatography-mass spectrometry (LC/MS) method we determined sex steroid (17ß-estradiol [E2], testosterone, androstenedione, and progesterone) fluctuations in serum and in 15 tissues throughout the murine estrous cycle (proestrus, estrus, and diestrus I) and in ovariectomized (OVX) mice. We observed dynamic fluctuations in serum and tissue steroid content throughout the estrous cycle with proestrus generally presenting the highest content of E2, testosterone, and androstenedione, and lowest content of progesterone. In general, the trend in circulating steroid content between the stages of the estrous cycle was mimicked in tissue. However, the absolute amounts of steroid levels when normalized to tissue weight were found to be significantly different between the tissues with the serum steroid quantity often being significantly lower than the tissue quantity. Additionally, we found that OVX mice generally displayed a depletion of all steroids in the various tissues assessed, except in the adrenal glands which were determined to be the main site of peripheral E2 production after ovary removal. This investigation provides a comprehensive analysis of steroid content throughout the estrous cycle in a multitude of tissues and serum. We believe this information will help serve as the basis for the development of physiologically relevant, tissue-specific hormonal therapies.


Assuntos
Androstenodiona , Progesterona , Feminino , Camundongos , Animais , Hormônios Esteroides Gonadais , Estradiol , Ciclo Estral/fisiologia , Testosterona
9.
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
10.
Nutrients ; 15(13)2023 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-37447375

RESUMO

Fibroblast growth factor 21 (FGF21), a hormone predominantly released in the liver, has emerged as a critical endocrine signal of dietary protein intake, but its role in the control of estrous cyclicity by dietary protein remains uncertain. To investigated the role of FGF21 and hypothalamic changes in the regulation of estrous cyclicity by dietary protein intake, female adult Sprague-Dawley rats with normal estrous cycles were fed diets with protein contents of 4% (P4), 8% (P8), 13% (P13), 18% (P18), and 23% (P23). FGF21 liver-specific knockout or wild-type mice were fed P18 or P4 diets to examine the role of liver FGF21 in the control of estrous cyclicity. Dietary protein restriction resulted in no negative effects on estrous cyclicity or ovarian follicular development when the protein content was greater than 8%. Protein restriction at 4% resulted in decreased bodyweight, compromised Kiss-1 expression in the hypothalamus, disturbed estrous cyclicity, and inhibited uterine and ovarian follicular development. The disturbed estrous cyclicity in rats that received the P4 diet was reversed after feeding with the P18 diet. Liver Fgf21 mRNA expressions and serum FGF21 levels were significantly increased as dietary protein content decreased, and loss of hepatic FGF21 delayed the onset of cyclicity disruption in rats fed with the P4 diet, possibly due to the regulation of insulin-like growth factor-1. Collectively, severe dietary protein restriction results in the cessation of estrous cyclicity and ovarian follicle development, and hepatic FGF21 and hypothalamic Kiss-1 were partially required for this process.


Assuntos
Proteínas na Dieta , Kisspeptinas , Ratos , Camundongos , Feminino , Animais , Proteínas na Dieta/farmacologia , Proteínas na Dieta/metabolismo , Kisspeptinas/metabolismo , Ratos Sprague-Dawley , Ciclo Estral/fisiologia , Fatores de Crescimento de Fibroblastos/metabolismo , Fígado/metabolismo
11.
Anim Reprod Sci ; 250: 107212, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36913896

RESUMO

Visfatin/NAMPT creates a hormonal link between energy metabolism and female reproduction. A recent study documented visfatin expression in the ovary and its action on follicular cells; however, the expression of visfatin in luteal cells is still unknown. The aim of this study, therefore, was to investigate the transcript and protein expression of visfatin as well as its immunolocalization in the corpus luteum (CL) and to examine the involvement of extracellular signal-regulated kinases (ERK1/2) in the regulation of visfatin level in response to LH, insulin, progesterone (P4), prostaglandin E2 (PGE2) and F2α (PGF2α). Corpora lutea were harvested from gilts on days 2-3, 10-12 and 14-16 of the estrous cycle and on days 10-11, 12-13, 15-16 and 27-28 of pregnancy. The current study demonstrated that visfatin expression depends on hormonal status related to the phase of the estrous cycle or early pregnancy. Visfatin was immunolocalized to the cytoplasm of small and large luteal cells. Moreover, visfatin protein abundance was increased by P4, and decreased by both prostaglandins, while LH and insulin have modulatory effects, depending on the phase of the cycle. Interestingly, LH, P4 and PGE2 effects were abolished in response to the inhibition of ERK1/2 kinase. Thus, this study demonstrated that expression of visfatin in the porcine CL is determined by the endocrine status related to the estrous cycle and early pregnancy and by the action of LH, insulin, P4 and prostaglandins via activation of the ERK1/2 pathway.


Assuntos
Insulinas , Nicotinamida Fosforribosiltransferase , Gravidez , Feminino , Suínos , Animais , Nicotinamida Fosforribosiltransferase/genética , Nicotinamida Fosforribosiltransferase/metabolismo , Nicotinamida Fosforribosiltransferase/farmacologia , Corpo Lúteo/fisiologia , Progesterona/metabolismo , Ciclo Estral/fisiologia , Prostaglandinas/metabolismo , Dinoprostona/metabolismo , Insulinas/metabolismo , Insulinas/farmacologia , Dinoprosta/farmacologia , Dinoprosta/metabolismo
12.
Curr Biol ; 33(7): 1358-1364.e4, 2023 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-36889318

RESUMO

Behavior is shaped by both the internal state of an animal and its individual behavioral biases. Rhythmic variation in gonadal hormones during the estrous cycle is a defining feature of the female internal state, one that regulates many aspects of sociosexual behavior. However, it remains unclear whether estrous state influences spontaneous behavior and, if so, how these effects might relate to individual behavioral variation. Here, we address this question by longitudinally characterizing the open-field behavior of female mice across different phases of the estrous cycle, using unsupervised machine learning to decompose spontaneous behavior into its constituent elements.1,2,3,4 We find that each female mouse exhibits a characteristic pattern of exploration that uniquely identifies it as an individual across many experimental sessions; by contrast, estrous state only negligibly impacts behavior, despite its known effects on neural circuits that regulate action selection and movement. Like female mice, male mice exhibit individual-specific patterns of behavior in the open field; however, the exploratory behavior of males is significantly more variable than that expressed by females both within and across individuals. These findings suggest underlying functional stability to the circuits that support exploration in female mice, reveal a surprising degree of specificity in individual behavior, and provide empirical support for the inclusion of both sexes in experiments querying spontaneous behaviors.


Assuntos
Ciclo Estral , Comportamento Exploratório , Camundongos , Masculino , Feminino , Animais , Ciclo Estral/fisiologia , Comportamento Exploratório/fisiologia , Movimento
13.
Physiol Behav ; 263: 114136, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36841322

RESUMO

According to the different stages of the estrous cycle, female rats exhibit behavioral changes associated with variations in sex hormone levels that affect the functionality of certain brain regions. In this study, we characterized the attention that female rats paid to a sexually-experienced male and the degree of electroencephalographic (EEG) activation and coupling between the medial prefrontal and posterior parietal cortices during antagonistic phases of the estrous cycle (proestrus-estrus vs. diestrous). The degree of attention paid to the stimulus was measured by the number of nose pokes performed while the rats were in a sexual incentive motivation box. EEGs were recorded in two conditions: a) awake-quiet state with no male rat present; and b) awake-quiet state in the presence of a male. Only during proestrus-estrus did the females show lower latency with a higher frequency and duration of nose pokes. In both cortices, the receptive females presented higher absolute power in all EEG bands recorded in the presence of the male, regardless of the phase of the estrous cycle. They also had greater EEG coupling between the medial prefrontal and posterior parietal cortices of the left hemisphere in all EEG bands regardless of the presence of a male. The higher synchronization between prefronto-parietal areas could be associated with the greater attention paid to, and adequate processing of, the sexual stimuli emitted by the male. Hence, it is probable that manifesting the proceptivity and receptivity behaviors characteristic of the proestrus-estrus phase requires a higher functional coupling between the prefrontal and parietal cortices.


Assuntos
Eletroencefalografia , Ciclo Estral , Masculino , Ratos , Feminino , Animais , Proestro , Ciclo Estral/fisiologia , Comportamento Sexual Animal/fisiologia , Estro/fisiologia
14.
Sci Rep ; 13(1): 2392, 2023 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-36765080

RESUMO

After ovulation, the mitochondrial enzyme CYP11A1 cleavage the cholesterol into pregnenolone for progesterone synthesis, suggesting that mitochondrial dynamics play a vital role in the female reproductive system. The changes in the mitochondria dynamics throughout the ovarian cycle have been reported in literature, but the correlation to its role in the ovarian cycle remains unclear. In this study, mitochondrial fusion promotor, M1, was used to study the impact of mitochondria dynamics in the female reproductive system. Our results showed that M1 treatment in mice can lead to the disruptions of estrous cycles in vagina smears. The decrease in serum LH was recorded in the animal. And the inhibitions of progesterone secretion and ovulations were observed in ovarian culture. Although no significant changes in mitochondrial networks were observed in the ovaries, significant up-regulation of mitochondrial respiratory complexes was revealed in M1 treatments through transcriptomic analysis. In contrast to the estrogen and steroid biosynthesis up-regulated in M1, the molecules of extracellular matrix, remodeling enzymes, and adhesion signalings were decreased. Collectively, our study provides novel targets to regulate the ovarian cycles through the mitochondria. However, more studies are still necessary to provide the functional connections between mitochondria and the female reproductive systems.


Assuntos
Dinâmica Mitocondrial , Progesterona , Camundongos , Feminino , Animais , Proestro , Ciclo Estral/fisiologia , Ovário , Estradiol
15.
Theriogenology ; 198: 332-343, 2023 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-36640738

RESUMO

Uterine secretions provide a suitable environment for sperm selective migration during a couple of days preceding ovulation and for early embryo development before implantation. Our goal was to identify and quantify proteins in the bovine uterine fluid during the periovulatory period of the estrous cycle. Genital tracts with normal morphology were collected from adult cyclic Bos taurus females in a local slaughterhouse and classified into pre-ovulatory or post-ovulatory stages of cycle (around days 19-21 and 0-5 of cycle, respectively; n = 8 cows per stage) based on ovarian morphology. Proteins from uterine fluid collected from the utero-tubal junction to the base of each horn (four pools of two cows per condition) were analyzed by nanoLiquid Chromatography coupled with tandem Mass Spectrometry (nanoLC-MS/MS). A total of 1214 proteins were identified, of which 91% were shared between all conditions. Overall, 57% of proteins were predicted to be secreted and 17% were previously reported in uterine extracellular vesicles. Paired comparisons between uterine horns ipsilateral and contralateral to ovulation evidenced 12 differentially abundant proteins, including five at pre-ovulatory stage. Furthermore, 35 proteins differed in abundance between pre- and post-ovulatory stages, including 21 in the ipsilateral side of ovulation. Functional analysis of identified proteins demonstrated roles in binding, metabolism, cellular detoxification and the immune response. This study provides a valuable database of uterine proteins for functional studies on sperm physiology and early embryo development.


Assuntos
Ovário , Proteoma , Feminino , Bovinos , Animais , Masculino , Ovário/metabolismo , Proteoma/metabolismo , Espectrometria de Massas em Tandem/veterinária , Sêmen/metabolismo , Ciclo Estral/fisiologia , Ovulação
16.
J Neurosci ; 43(5): 736-748, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36549906

RESUMO

The estrous cycle is a potent modulator of neuron physiology. In rodents, in vivo ventral tegmental area (VTA) dopamine (DA) activity has been shown to fluctuate across the estrous cycle. Although the behavioral effect of fluctuating sex steroids on the reward circuit is well studied in response to drugs of abuse, few studies have focused on the molecular adaptations in the context of stress and motivated social behaviors. We hypothesized that estradiol fluctuations across the estrous cycle acts on the dopaminergic activity of the VTA to alter excitability and stress response. We used whole-cell slice electrophysiology of VTA DA neurons in naturally cycling, adult female C57BL/6J mice to characterize the effects of the estrous cycle and the role of 17ß-estradiol on neuronal activity. We show that the estrous phase alters the effect of 17ß-estradiol on excitability in the VTA. Behaviorally, the estrous phase during a series of acute variable social stressors modulates subsequent reward-related behaviors. Pharmacological inhibition of estrogen receptors in the VTA before stress during diestrus mimics the stress susceptibility found during estrus, whereas increased potassium channel activity in the VTA before stress reverses stress susceptibility found during estrus as assessed by social interaction behavior. This study identifies one possible potassium channel mechanism underlying the increased DA activity during estrus and reveals estrogen-dependent changes in neuronal function. Our findings demonstrate that the estrous cycle and estrogen signaling changes the physiology of DA neurons resulting in behavioral differences when the reward circuit is challenged with stress.SIGNIFICANCE STATEMENT The activity of the ventral tegmental area encodes signals of stress and reward. Dopaminergic activity has been found to be regulated by both local synaptic inputs as well as inputs from other brain regions. Here, we provide evidence that cycling sex steroids also play a role in modulating stress sensitivity of dopaminergic reward behavior. Specifically, we reveal a correlation of ionic activity with estrous phase, which influences the behavioral response to stress. These findings shed new light on how estrous cycle may influence dopaminergic activity primarily during times of stress perturbation.


Assuntos
Neurônios Dopaminérgicos , Ciclo Estral , Camundongos , Animais , Feminino , Camundongos Endogâmicos C57BL , Neurônios Dopaminérgicos/fisiologia , Ciclo Estral/fisiologia , Estrogênios/farmacologia , Estradiol/farmacologia , Comportamento Social , Mesencéfalo , Canais de Potássio , Área Tegmentar Ventral
17.
Psychopharmacology (Berl) ; 240(1): 15-25, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36571628

RESUMO

RATIONALE: The female menstrual or estrous cycle and its associated fluctuations in circulating estradiol (E2), progesterone, and other gonadal hormones alter orexin or hypocretin peptide production and receptor activity. Depending on the estrous cycle phase, the transcription of prepro-orexin mRNA, post-translational modification of orexin peptide, and abundance of orexin receptors change in a brain region-specific manner. The most dramatic changes occur in the hypothalamus, which is considered the starting point of the hypothalamic-pituitary-gonadal axis as well as the hub of orexin-producing neurons. Thus, hypothalamus-regulated behaviors, including arousal, feeding, reward processing, and the stress response depend on coordinated efforts between E2, progesterone, and the orexin system. Given the rise of orexin therapeutics for various neuropsychiatric conditions including insomnia and affective disorders, it is important to delineate the behavioral outcomes of this drug class in both sexes, as well as within different time points of the female reproductive cycle. OBJECTIVES: Summarize how the menstrual or estrous cycle affects orexin system functionality in animal models in order to predict how orexin pharmacotherapies exert varying degrees of behavioral effects across the dynamic hormonal milieu.


Assuntos
Estradiol , Progesterona , Masculino , Animais , Feminino , Orexinas , Receptores de Orexina , Peptídeos , Ciclo Estral/fisiologia
18.
Am J Physiol Heart Circ Physiol ; 323(6): H1057-H1067, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36240435

RESUMO

With the increasing appreciation for sex as a biological variable and the inclusion of female mice in research, it is important to understand the influence of the estrous cycle on physiological function. Sex hormones are known to modulate vascular function, but the effects of the mouse estrous cycle phase on arterial stiffness, endothelial function, and arterial estrogen receptor expression remain unknown. In 23 female C57BL/6 mice (6 mo of age), we determined the estrous cycle stage via vaginal cytology and plasma hormone concentrations. Aortic stiffness, assessed by pulse wave velocity, was lower during the estrus phase compared with diestrus. In ex vivo assessment of isolated pressurized mesenteric and posterior cerebral arteries, the responses to acetylcholine, insulin, and sodium nitroprusside, as well as nitric oxide-mediated dilation, were not different between estrous cycle phases. In the aorta, expression of phosphorylated estrogen receptor-α was higher for mice in estrus compared with mice in proestrus. In the cerebral arteries, gene expression for estrogen receptor-ß (Esr2) was lowest for mice in estrus compared with diestrus and proestrus. These results demonstrate that the estrus phase is associated with lower in vivo large artery stiffness in mice. In contrast, ex vivo resistance artery endothelial function is not different between estrous cycle phases. Estrogen receptor expression is modulated by the estrus cycle in an artery-dependent manner. These results suggest that the estrous cycle phase should be considered when measuring in vivo arterial stiffness in young female mice.NEW & NOTEWORTHY To design rigorous vascular research studies using young female rodents, the influence of the estrous cycle on vascular function must be known. We found that in vivo aortic stiffness was lower during estrus compared with the diestrus phase in female mice. In contrast, ex vivo mesenteric and cerebral artery endothelial function did not differ between estrous cycle stages. These results suggest that the estrous cycle stage should be accounted for when measuring in vivo arterial stiffness.


Assuntos
Rigidez Vascular , Camundongos , Feminino , Animais , Receptores de Estrogênio , Análise de Onda de Pulso , Camundongos Endogâmicos C57BL , Ciclo Estral/fisiologia , Estrogênios , Artérias
19.
Elife ; 112022 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-36205477

RESUMO

The estrous cycle is regulated by rhythmic endocrine interactions of the nervous and reproductive systems, which coordinate the hormonal and ovulatory functions of the ovary. Folliculogenesis and follicle progression require the orchestrated response of a variety of cell types to allow the maturation of the follicle and its sequela, ovulation, corpus luteum formation, and ovulatory wound repair. Little is known about the cell state dynamics of the ovary during the estrous cycle and the paracrine factors that help coordinate this process. Herein, we used single-cell RNA sequencing to evaluate the transcriptome of >34,000 cells of the adult mouse ovary and describe the transcriptional changes that occur across the normal estrous cycle and other reproductive states to build a comprehensive dynamic atlas of murine ovarian cell types and states.


Assuntos
Ovário , Ovulação , Animais , Ciclo Estral/fisiologia , Feminino , Camundongos , Folículo Ovariano/fisiologia , Ovulação/fisiologia , Pelve
20.
J Equine Vet Sci ; 118: 104131, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36184019

RESUMO

After having been brought to America 400 years ago, the Criollo horse reproduced freely for centuries in the southern part of the American continent. Roughness, resistance, and endurance are typical characteristics of this breed of horses. Although the natural selection that occurred over the centuries may have positively influenced the fertility and longevity of this breed, information regarding ovarian function and other reproductive aspects of Criollo breed mares under natural or controlled management conditions is scarce. The objectives of the present study were to (1) characterize the follicular dynamics of Criollo breed mares, (2) characterize growth and regression of the corpus luteum, and (3) study the cervical and uterine tone and ultrasonographic echotexture changes during two consecutive estrous cycles. In this study, 26 interovulatory intervals (IOI) were evaluated in 13 mares. Spontaneous estrous cycles were characterized by the following: (1) the IOI length was 21 days; (2) dominant and subordinate follicles emerged together at day 5 (ovulation = day 0), and their growth rates were slower until the day of deviation; (3) the deviation in growth rates between the two largest follicles of the ovulatory wave occurred when the dominant and subordinate follicles reached 22 and 21 mm in diameter, respectively; (4) the mean diameter of the preovulatory follicle at the day before ovulation was 43 mm; (5) different combinations of follicular waves were observed in each interovulatory interval; (6) the corpus luteum reached its maximum diameter about 2 days after ovulation and decreased gradually afterward; (7) after ovulation, the cervical and uterine tones were positively correlated and remained elevated until the onset of the expected luteolysis (days 12-14); and (8) after the expected luteolysis, the endometrial echotexture started to increase and reached maximum values 5-3 days before ovulation, when it started to decrease. Moderate-to-strong positive correlations between IOIs revealed repeatability within animals for the diameter of the preovulatory follicle at maximum and at the day before ovulation, cervical and uterine tones, endometrial echotexture, and corpus luteum diameter. The findings herein presented are of fundamental importance for a better understanding of the reproductive physiological patterns of the estrous cycle in the Criollo breed mare.


Assuntos
Ciclo Estral , Ovulação , Cavalos , Feminino , Animais , Ciclo Estral/fisiologia , Ovulação/fisiologia , Corpo Lúteo/diagnóstico por imagem , Ovário/fisiologia , Folículo Ovariano/diagnóstico por imagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...