Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.146
Filtrer
1.
Int J Mol Sci ; 25(17)2024 Aug 30.
Article de Anglais | MEDLINE | ID: mdl-39273375

RÉSUMÉ

Determining the estrous cycle stages in mice is essential for optimizing breeding strategies, synchronizing experimental timelines, and facilitating studies in behavior, drug testing, and genetics. It is critical for reducing the production of genetically unmodified offspring in the generation and investigation of genetically modified animal models. An accurate detection of the estrus cycle is particularly relevant in the context of the 3Rs-Replacement, Reduction, and Refinement. The estrous cycle, encompassing the reproductive phases of mice, is key to refining experimental designs and addressing ethical issues related to the use of animals in research. This study presents results from two independent laboratories on the efficacy of the Mouse Estrus Detector (MED) from ELMI Ltd. (Latvia) for the accurate determination of the estrus phase. The female mice of five strains/stocks (CD1, FVB/N, C57Bl6/J, B6D2F1, and Swiss) were used. The results showed that the MEDProTM is a low-traumatic, simple, rapid, and painless method of estrus detection that supports the principles of the 3Rs. The use of the MEDProTM for estrus detection in mice caused minimal stress, enhanced mating efficiency, facilitated an increase in the number of embryos for in vitro fertilization, and allowed the production of the desired number of foster animals.


Sujet(s)
Détection de l'oestrus , Oestrus , Animaux , Souris , Femelle , Oestrus/physiologie , Détection de l'oestrus/méthodes , Cycle oestral/physiologie , Mâle
2.
BMC Vet Res ; 20(1): 401, 2024 Sep 09.
Article de Anglais | MEDLINE | ID: mdl-39245728

RÉSUMÉ

Successful identification of estrum or other stages in a cycling bitch often requires a combination of methods, including assessment of its behavior, exfoliative vaginal cytology, vaginoscopy, and hormonal assays. Vaginoscopy is a handy and inexpensive tool for the assessment of the breeding period. The present study introduces an innovative method for identifying the stages in the estrous cycle of female canines. With a dataset of 210 vaginoscopic images covering four reproductive stages, this approach extracts deep features using the inception v3 and Residual Networks (ResNet) 152 models. Binary gray wolf optimization (BGWO) is applied for feature optimization, and classification is performed with the extreme gradient boosting (XGBoost) algorithm. Both models are compared with the support vector machine (SVM) with the Gaussian and linear kernel, k-nearest neighbor (KNN), and convolutional neural network (CNN), based on performance metrics such as accuracy, specificity, F1 score, sensitivity, precision, matthew correlation coefficient (MCC), and runtime. The outcomes demonstrate the superiority of the deep model of ResNet 152 with XGBoost classifier, achieving an average model accuracy of 90.37%. The method gave a specific accuracy of 90.91%, 96.38%, 88.37%, and 88.24% in predicting the proestrus, estrus, diestrus, and anestrus stages, respectively. When performing deep feature analysis using inception v3 with the same classifiers, the model achieved an accuracy of 89.41%, which is comparable to the results obtained with the ResNet model. The proposed model offers a reliable system for identifying the optimal mating period, providing breeders and veterinarians with an efficient tool to enhance the success of their breeding programs.


Sujet(s)
Apprentissage profond , Animaux , Femelle , Chiens , Cycle oestral/physiologie , Vagin , Machine à vecteur de support , Oestrus/physiologie
3.
Cell Tissue Res ; 397(3): 275-285, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39105776

RÉSUMÉ

The complex interactome crucial for successful pregnancy is constituted by the intricate network of endocrine and paracrine signaling pathways, involving gametes, embryos, and the female reproductive tract. Specifically, the oviduct exhibits distinct responses to gametes and early embryos during particular phases of the estrus cycle, a process tightly regulated by reproductive hormones. Moreover, these hormones play a pivotal role in orchestrating cyclical changes within oviductal epithelial cells. To unravel the molecular mechanisms underlying these dynamic changes, our study aimed to investigate the involvement of protein kinase A (PKA) in oviductal epithelial cells throughout the estrus cycle and in advanced pregnancy, extending our studies to oviductal epithelial cell in primary culture. By a combination of 2D-gel electrophoresis, Western blotting, and mass spectrometry, we identified 17 proteins exhibiting differential phosphorylation status mediated by PKA. Among these proteins, we successfully validated the phosphorylation status of heat shock 70 kDa protein (HSP70), aconitase 2 (ACO2), and lamin B1 (LMNB1). Our findings unequivocally demonstrate the dynamic regulation of PKA throughout the estrus cycle in oviductal epithelial cells. Also, analysis by bioinformatics tools suggest its pivotal role in mediating cyclical changes possibly through modulation of apoptotic pathways. This research sheds light on the intricate molecular mechanisms underlying reproductive processes, with implications for understanding fertility and reproductive health.


Sujet(s)
Apoptose , Cyclic AMP-Dependent Protein Kinases , Cellules épithéliales , Cycle oestral , Transduction du signal , Animaux , Femelle , Cellules épithéliales/métabolisme , Bovins , Cyclic AMP-Dependent Protein Kinases/métabolisme , Cycle oestral/physiologie , Cycle oestral/métabolisme , Oviductes/métabolisme , Oviductes/cytologie , Trompes utérines/métabolisme , Trompes utérines/cytologie , Phosphorylation
4.
J Neurophysiol ; 132(2): 403-417, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39106208

RÉSUMÉ

Cholinergic interneurons (ChIs) provide the main source of acetylcholine in the striatum and have emerged as a critical modulator of behavioral flexibility, motivation, and associative learning. In the dorsal striatum (DS), ChIs display heterogeneous firing patterns. Here, we investigated the spontaneous firing patterns of ChIs in the nucleus accumbens (NAc) shell, a region of the ventral striatum. We identified four distinct ChI firing signatures: regular single-spiking, irregular single-spiking, rhythmic bursting, and a mixed-mode pattern composed of bursting activity and regular single spiking. ChIs from females had lower firing rates compared with males and had both a higher proportion of mixed-mode firing patterns and a lower proportion of regular single-spiking neurons compared with males. We further observed that across the estrous cycle, the diestrus phase was characterized by higher proportions of irregular ChI firing patterns compared with other phases. Using pooled data from males and females, we examined how the stress-associated neuropeptide corticotropin releasing factor (CRF) impacts these firing patterns. ChI firing patterns showed differential sensitivity to CRF. This translated into differential ChI sensitivity to CRF across the estrous cycle. Furthermore, CRF shifted the proportion of ChI firing patterns toward more regular spiking activity over bursting patterns. Finally, we found that repeated stressor exposure altered ChI firing patterns and sensitivity to CRF in the NAc core, but not the NAc shell. These findings highlight the heterogeneous nature of ChI firing patterns, which may have implications for accumbal-dependent motivated behaviors.NEW & NOTEWORTHY Cholinergic interneurons (ChIs) within the dorsal and ventral striatum can exert a major influence on network output and motivated behaviors. However, the firing patterns and neuromodulation of ChIs within the ventral striatum, specifically the nucleus accumbens (NAc) shell, are understudied. Here, we report that NAc shell ChIs have heterogeneous ChI firing patterns that are labile and can be modulated by the stress-linked neuropeptide corticotropin releasing factor (CRF) and by the estrous cycle.


Sujet(s)
Neurones cholinergiques , Corticolibérine , Interneurones , Noyau accumbens , Animaux , Corticolibérine/métabolisme , Corticolibérine/pharmacologie , Femelle , Mâle , Interneurones/physiologie , Interneurones/métabolisme , Noyau accumbens/physiologie , Noyau accumbens/métabolisme , Noyau accumbens/cytologie , Neurones cholinergiques/physiologie , Neurones cholinergiques/métabolisme , Cycle oestral/physiologie , Potentiels d'action/physiologie , Souris
5.
Alzheimers Res Ther ; 16(1): 183, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39143583

RÉSUMÉ

BACKGROUND: Alzheimer's disease (AD) is the most prevalent dementia, showing higher incidence in women. Besides, lipids play an essential role in brain, and they could be dysregulated in neurodegeneration. Specifically, impaired plasma lipid levels could predict early AD diagnosis. This work aims to identify the main plasma lipids altered in early AD female mouse model and evaluate their relationship with brain lipidome. Also, the possible involvement of the estrous cycle in lipid metabolism has been evaluated. METHODS: Plasma samples of wild-type (n = 10) and APP/PS1 (n = 10) female mice of 5 months of age were collected, processed, and analysed using a lipidomic mass spectrometry-based method. A statistical analysis involving univariate and multivariate approaches was performed to identify significant lipid differences related to AD between groups. Also, cytology tests were conducted to confirm estrous cycle phases. RESULTS: Three hundred thirty lipids were detected in plasma, 18 of them showed significant differences between groups; specifically, some triacylglycerols, cholesteryl esters, lysophosphatidylcholines, phosphatidylcholines, and ether-linked phosphatidylcholines, increased in early AD; while other phosphatidylcholines, phosphatidylethanolamines, ceramides, and ether-linked phosphatidylethanolamines decreased in early AD. A multivariate approach was developed from some lipid variables, showing high diagnostic indexes (70% sensitivity, 90% specificity, 80% accuracy). From brain and plasma lipidome, some significant correlations were observed, mainly in the glycerophospholipid family. Also, some differences were found in both plasma and brain lipids, according to the estrous cycle phase. CONCLUSIONS: Therefore, lipid alterations can be identified in plasma at early AD stages in mice females, with a relationship with brain lipid metabolism for most of the lipid subfamilies, suggesting some lipids as potential AD biomarkers. In addition, the estrous cycle monitoring could be relevant in female studies.


Sujet(s)
Maladie d'Alzheimer , Précurseur de la protéine bêta-amyloïde , Encéphale , Modèles animaux de maladie humaine , Cycle oestral , Lipidomique , Lipides , Souris transgéniques , Animaux , Femelle , Cycle oestral/physiologie , Cycle oestral/sang , Lipidomique/méthodes , Maladie d'Alzheimer/sang , Maladie d'Alzheimer/métabolisme , Encéphale/métabolisme , Précurseur de la protéine bêta-amyloïde/métabolisme , Précurseur de la protéine bêta-amyloïde/sang , Précurseur de la protéine bêta-amyloïde/génétique , Lipides/sang , Préséniline-1/génétique , Souris , Métabolisme lipidique/physiologie , Souris de lignée C57BL
6.
Reprod Domest Anim ; 59(8): e14692, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39091039

RÉSUMÉ

Anti-Müllerian hormone (AMH) is a hormone produced by growing preantral and antral follicles of the ovary. AMH is accepted as an important biomarker for fertility and superovulation parameters in livestock species. This study aimed to evaluate changes in serum AMH level in the oestrous cycle, repeatability of AMH, the effect of age on serum AMH level and the effects of AMH on litter size in Romanov sheep. In the study, a total of 36 Romanov sheep were used as animal material. First blood samples (0th day) were collected from 36 ewes to evaluate AMH and progesterone levels. Second blood samples were collected randomly from 20 ewes 9 days after first sampling to compare AMH levels at two different periods of the oestrous cycle in Romanov ewes. The ewes were categorized into three groups as low, medium and high AMH based on their first AMH levels. Results indicated that serum AMH level did not change during the oestrous and dioestrous phases of the oestrous cycle and two random time points of the oestrous cycle (p > .05). Pearson correlation analysis revealed that there is a high (r = .95) and significant (p < .001) correlation between AMH levels at the 0th (AMH-1) and 9th (AMH-2) days. The effect of AMH level on litter size was found to be significant. Litter size was significantly higher in the high AMH group than in the low AMH group (p < .05). In addition, the age of ewes did not affect serum AMH levels (p > .05). ROC analysis indicates that AMH cut-off value >320 pg/mL with 70% sensitivity and 100% specificity can be used for litter size in Romanov ewes. In conclusion, AMH is highly repeatable and its serum AMH level did not change during the oestrous cycle in Romanov sheep. In addition, AMH affects litter size and can be reliably used as a marker for litter size in Romanov sheep.


Sujet(s)
Hormone antimullérienne , Marqueurs biologiques , Taille de la portée , Progestérone , Animaux , Hormone antimullérienne/sang , Femelle , Marqueurs biologiques/sang , Progestérone/sang , Cycle oestral/sang , Cycle oestral/physiologie , Ovis aries/physiologie , Ovis/physiologie
7.
Theriogenology ; 228: 110-120, 2024 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-39141998

RÉSUMÉ

Successful reproductive management of domestic mammals depends primarily upon timely identification of oestrous cycle stages. There is a need to develop an alternative non-invasive, welfare-friendly, accurate and reliable method to identify reproductive cycle stages. This is of particular interest for horse breeders, because horses are high-value farm animals that require careful management and individual monitoring. Saliva sampling is non-invasive, painless and welfare-friendly. Thus, we performed a metabolomic analysis of equine saliva during different reproductive stages to identify changes in the salivary metabolome during anoestrus, the oestrous cycle and early gestation. We compared the saliva and plasma metabolomes to investigate the relationship between the two fluids according to the physiological stage. We collected saliva and plasma samples from six mares during seasonal anoestrus, during the follicular phase 3 days, 2 days and 1 day before ovulation and the day when ovulation was detected, during the luteal phase 6 days after ovulation, and during early gestation 18 days after ovulation and insemination. Metabolome analysis was performed by proton-nuclear magnetic resonance spectroscopy. We identified 58 and 51 metabolites in saliva and plasma, respectively. The levels of four metabolites or groups of metabolites in saliva and five metabolites or groups of metabolites in plasma showed significant modifications during the 4 days until ovulation, ie 3 days prior to and on the day of ovulation. The levels of 11 metabolites or groups of metabolites in saliva and 17 metabolites or groups of metabolites in plasma were significantly different between the seasonal anoestrus and the ovarian cyclicity period. The physiological mechanisms involved in the onset of ovarian cyclicity and in ovulation induced modifications of the metabolome both in plasma and saliva. The metabolites whose salivary levels changed during the reproductive cycle could be potential salivary biomarkers to detect the reproductive stage in a welfare friendly production system. In particular, we propose creatine and alanine as candidate salivary biomarkers of ovulation and of the onset of ovarian cyclicity, respectively. However, extensive validation of their reliability is required. Our study contributes to extend to domestic mammals the use of saliva as a non-invasive alternative diagnostic fluid for reproduction in a welfare-friendly production system.


Sujet(s)
Anoestrus , Cycle oestral , Métabolome , Gestation animale , Salive , Animaux , Femelle , Salive/composition chimique , Salive/métabolisme , Equus caballus/physiologie , Equus caballus/métabolisme , Equus caballus/sang , Métabolome/physiologie , Grossesse , Cycle oestral/métabolisme , Cycle oestral/physiologie , Cycle oestral/sang , Projets pilotes , Gestation animale/métabolisme , Gestation animale/sang , Anoestrus/métabolisme , Anoestrus/physiologie
8.
Reprod Domest Anim ; 59(8): e14710, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39169649

RÉSUMÉ

The reproductive efficiency in buffalo is highly influenced by seasonal variability. Angiogenesis in the reproductive cycle is important for optimal physiological functioning of uterus. Estrogen receptor-α (ERα), vascular endothelial growth factor (VEGF) and reduced nicotinamide adenine dinucleotide phosphatase diaphorase (NADPH-d) are vital indicators for the uterine angiogenic process. This study was conducted to see the effect of season on the expression of different uterine angiogenic factors. Season wise (winter and summer) and phase wise (follicular and luteal), immune staining intensity of buffalo uterus was measured by calculating the optical density value (OD) for ERα and VEGF. Percentage of immuno-positive cell count for ERα was done. Histoenzymic NADPH-d expression was analysed. Expression of all these factors increased during follicular phase of oestrous cycle in order to support the angiogenesis; however, the expression was significantly lower (p ≤ 0.05) in term of OD value as well as percentage count of immuno-positive cells during summer season indicating lower angiogenic activity that subsequently affected reproduction in buffalo.


Sujet(s)
Buffles , Cycle oestral , Saisons , Utérus , Facteur de croissance endothéliale vasculaire de type A , Animaux , Buffles/métabolisme , Femelle , Utérus/métabolisme , Cycle oestral/métabolisme , Cycle oestral/physiologie , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Facteur de croissance endothéliale vasculaire de type A/génétique , Récepteur alpha des oestrogènes/métabolisme , Récepteur alpha des oestrogènes/génétique , Néovascularisation physiologique/physiologie
9.
Int J Biol Sci ; 20(10): 3863-3880, 2024.
Article de Anglais | MEDLINE | ID: mdl-39113716

RÉSUMÉ

The mechanisms behind the selection and initial recruitment of primordial follicles (PmFs) from the non-growing PmF pool during each estrous cycle in females remain largely unknown. This study demonstrates that PmFs closest to the ovulatory follicle are preferentially activated in mouse ovaries under physiological conditions. PmFs located within 40 µm of the ovulatory follicles were more likely to be activated compared to those situated further away during the peri-ovulation period. Repeated superovulation treatments accelerated the depletion of the PmF reserve, whereas continuous suppression of ovulation delayed PmF reserve consumption. Spatial transcriptome sequencing of peri-ovulatory follicles revealed that ovulation primarily induces the degradation and remodeling of the extracellular matrix (ECM). This ECM degradation reduces mechanical stress around PmFs, thereby triggering their activation. Specifically, Cathepsin L (CTSL), a cysteine proteinase and lysosomal enzyme involved in ECM degradation, initiates the activation of PmFs adjacent to ovulatory follicles in a distance-dependent manner. These findings highlight the link between ovulation and selective PmF activation, and underscore the role of CTSL in this process under physiological conditions.


Sujet(s)
Cathepsine L , Matrice extracellulaire , Follicule ovarique , Ovulation , Animaux , Femelle , Souris , Follicule ovarique/métabolisme , Cathepsine L/métabolisme , Ovulation/physiologie , Matrice extracellulaire/métabolisme , Ovaire/métabolisme , Cycle oestral/physiologie
10.
Metabolism ; 158: 155976, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39019342

RÉSUMÉ

BACKGROUND: Estrogen secretion by the ovaries regulates the hypothalamic-pituitary-gonadal axis during the reproductive cycle, influencing gonadotropin-releasing hormone (GnRH) and luteinizing hormone (LH) secretion, and also plays a role in regulating metabolism. Here, we establish that hypothalamic tanycytes-specialized glia lining the floor and walls of the third ventricle-integrate estrogenic feedback signals from the gonads and couple reproduction with metabolism by relaying this information to orexigenic neuropeptide Y (NPY) neurons. METHODS: Using mouse models, including mice floxed for Esr1 (encoding estrogen receptor alpha, ERα) and those with Cre-dependent expression of designer receptors exclusively activated by designer drugs (DREADDs), along with viral-mediated, pharmacological and indirect calorimetric approaches, we evaluated the role of tanycytes and tanycytic estrogen signaling in pulsatile LH secretion, cFos expression in NPY neurons, estrous cyclicity, body-weight changes and metabolic parameters in adult females. RESULTS: In ovariectomized mice, chemogenetic activation of tanycytes significantly reduced LH pulsatile release, mimicking the effects of direct NPY neuron activation. In intact mice, tanycytes were crucial for the estrogen-mediated control of GnRH/LH release, with tanycytic ERα activation suppressing fasting-induced NPY neuron activation. Selective knockout of Esr1 in tanycytes altered estrous cyclicity and fertility in female mice and affected estrogen's ability to inhibit refeeding in fasting mice. The absence of ERα signaling in tanycytes increased Npy transcripts and body weight in intact mice and prevented the estrogen-mediated decrease in food intake as well as increase in energy expenditure and fatty acid oxidation in ovariectomized mice. CONCLUSIONS: Our findings underscore the pivotal role of tanycytes in the neuroendocrine coupling of reproduction and metabolism, with potential implications for its age-related deregulation after menopause. SIGNIFICANCE STATEMENT: Our investigation reveals that tanycytes, specialized glial cells in the brain, are key interpreters of estrogen signals for orexigenic NPY neurons in the hypothalamus. Disrupting tanycytic estrogen receptors not only alters fertility in female mice but also impairs the ability of estrogens to suppress appetite. This work thus sheds light on the critical role played by tanycytes in bridging the hormonal regulation of cyclic reproductive function and appetite/feeding behavior. This understanding may have potential implications for age-related metabolic deregulation after menopause.


Sujet(s)
Cellules épendymogliales , Récepteur alpha des oestrogènes , Fécondité , Hormone lutéinisante , Transduction du signal , Animaux , Récepteur alpha des oestrogènes/métabolisme , Récepteur alpha des oestrogènes/génétique , Femelle , Souris , Fécondité/physiologie , Cellules épendymogliales/métabolisme , Transduction du signal/physiologie , Hormone lutéinisante/métabolisme , Cycle oestral/physiologie , Cycle oestral/métabolisme , Neuropeptide Y/métabolisme , Ovariectomie , Neurones/métabolisme , Hypothalamus/métabolisme , Souris de lignée C57BL , Hormone de libération des gonadotrophines/métabolisme
11.
J Biol Rhythms ; 39(5): 413-422, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39082411

RÉSUMÉ

The estrous cycle regulates rhythms of locomotor activity, body temperature, and circadian gene expression. In female mice, activity increases on the night of proestrus, when elevated estrogens cause ovulation. Exogenous estradiol regulates eating behavior rhythms in female mice fed a high-fat diet, but it is unknown whether endogenous estrogens regulate eating rhythms. In this study, we investigated whether diurnal and circadian eating behavior rhythms change systematically across the estrous cycle. We first studied diurnal eating behavior rhythms in female C57BL/6J mice in 12L:12D. Estrous cycle stages were determined by vaginal cytology while eating behavior and wheel revolutions were continuously measured. The mice had regular 4- to 5-day estrous cycles. Consistent with prior studies, the greatest number of wheel revolutions occurred on the night of proestrus into estrus when systemic levels of estrogens peak. The amplitude, or robustness, of the eating behavior rhythm also fluctuated with 4- to 5-day cycles and peaked primarily during proestrus or estrus. The phases of eating behavior rhythms fluctuated, but not at 4- or 5-day intervals, and phases did not correlate with estrous cycle stages. After ovariectomy, the eating behavior rhythm amplitude fluctuated at irregular intervals. In constant darkness, the amplitude of the circadian eating behavior rhythm peaked every 4 or 5 days and coincided with the circadian day that had the greatest number of wheel revolutions, a marker of proestrus. These data suggest that fluctuations of ovarian hormones across the estrous cycle temporally organize the robustness of circadian eating behavior rhythms so that it peaks during ovulation and sexual receptivity.


Sujet(s)
Rythme circadien , Cycle oestral , Comportement alimentaire , Souris de lignée C57BL , Animaux , Femelle , Cycle oestral/physiologie , Comportement alimentaire/physiologie , Souris , Activité motrice , Ovariectomie , Photopériode , Oestrogènes , Oestradiol
12.
Neuroscience ; 554: 118-127, 2024 Aug 30.
Article de Anglais | MEDLINE | ID: mdl-39019393

RÉSUMÉ

Despite significant advances in the study of fear and fear memory formation, little is known about fear learning and expression in females. This omission has been proven surprising, as normal and pathological behaviors are highly influenced by ovarian hormones, particularly estradiol and progesterone. In the current study, we investigated the joint influence of serotonin (5-HT) neurotransmission and estrous cycle phases (low or high levels of estradiol and progesterone) on the expression of conditioned fear in a group of female rats that were previously divided according to their response to stressful stimuli into low or high anxiety-like subjects. The baseline amplitude of the unconditioned acoustic startle responses was high in high-anxiety female rats, with no effect on the estrous cycle observed. Data collected during the proestrus-estrus phase revealed that low-anxiety rats had startle amplitudes similar to those of high-anxiety rats. It is supposed that high-anxiety female rats benefit from increased estradiol and progesterone levels to achieve comparable potentiated startle amplitudes. In contrast, female rats experienced a significant decrease in hormone levels during the Diestrus phase. This decrease is believed to play a role in preventing them from displaying a heightened startle response when faced with strongly aversive stimuli. Data collected after 5-HT and 8-OH-DPAT were administered into the basolateral nuclei and dorsal periaqueductal gray suggest that 5-HT neurotransmission works with progesterone and estrogen to reduce startle potentiation, most likely by activating the serotonin-1A receptor subtype.


Sujet(s)
Groupe nucléaire basolatéral , Oestradiol , Peur , Substance grise centrale du mésencéphale , Progestérone , Récepteur de la sérotonine de type 5-HT1A , Réflexe de sursaut , Animaux , Femelle , Rats , Anxiété/métabolisme , Anxiété/physiopathologie , Groupe nucléaire basolatéral/métabolisme , Groupe nucléaire basolatéral/effets des médicaments et des substances chimiques , Conditionnement classique/physiologie , Conditionnement classique/effets des médicaments et des substances chimiques , Oestradiol/pharmacologie , Oestradiol/métabolisme , Cycle oestral/physiologie , Peur/physiologie , Peur/effets des médicaments et des substances chimiques , Substance grise centrale du mésencéphale/métabolisme , Substance grise centrale du mésencéphale/effets des médicaments et des substances chimiques , Progestérone/pharmacologie , Progestérone/métabolisme , Rat Wistar , Récepteur de la sérotonine de type 5-HT1A/métabolisme , Réflexe de sursaut/physiologie , Réflexe de sursaut/effets des médicaments et des substances chimiques , Sérotonine/métabolisme
13.
Neurosci Biobehav Rev ; 164: 105789, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39002829

RÉSUMÉ

Anxiety fluctuates across the human menstrual cycle, with symptoms worsening during phases of declining or low ovarian hormones. Similar findings have been observed across the rodent estrous cycle, however, the magnitude and robustness of these effects have not been meta-analytically quantified. We conducted a systematic review and meta-analysis of estrous cycle effects on anxiety-like behaviour (124 articles; k = 259 effect sizes). In both rats and mice, anxiety-like behaviour was higher during metestrus/diestrus (lower ovarian hormones) than proestrus (higher ovarian hormones) (g = 0.44 in rats, g = 0.43 in mice). There was large heterogeneity in the data, which was partially accounted for by strain, experimental task, and reproductive status. Nonetheless, the effect of estrous cycle on anxiety-like behaviour was highly robust, with the fail-safe N test revealing the effect would remain significant even if 21,388 additional studies yielded null results. These results suggest that estrous cycle should be accounted for in studies of anxiety in females. Doing so will facilitate knowledge about menstrual-cycle regulation of anxiety disorders in humans.


Sujet(s)
Anxiété , Cycle oestral , Peur , Animaux , Femelle , Cycle oestral/physiologie , Anxiété/physiopathologie , Rats , Peur/physiologie , Souris , Comportement animal/physiologie
14.
BMC Vet Res ; 20(1): 315, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39010076

RÉSUMÉ

BACKGROUND: While the urogenital microbiota has recently been characterized in healthy male and female dogs, the influence of sex hormones on the urogenital microbiome of bitches is still unknown. A deeper understanding of the cyclic changes in urinary and vaginal microbiota would allow us to compare the bacterial populations in healthy dogs and assess the impact of the microbiome on various urogenital diseases. Therefore, the aim of this study was to characterize and compare the urogenital microbiota during different phases of the estrous cycle in healthy female dogs. DNA extraction, 16 S rDNA library preparation, sequencing and informatic analysis were performed to determine the vaginal and urinary microbiota in 10 healthy beagle dogs at each phase of the estrous cycle. RESULTS: There were no significant differences in alpha and beta diversity of the urinary microbiota across the different cycle phases. Similarly, alpha diversity, richness and evenness of vaginal bacterial populations were not significantly different across the cycle phases. However, there were significant differences in vaginal beta diversity between the different cycle phases, except for between anestrus and diestrus. CONCLUSION: This study strongly suggests that estrogen influences the abundance of the vaginal microbiota in healthy female dogs, but does not appear to affect the urinary microbiome. Furthermore, our data facilitate a deeper understanding of the native urinary and vaginal microbiota in healthy female dogs.


Sujet(s)
Cycle oestral , Microbiote , Vagin , Animaux , Chiens , Femelle , Vagin/microbiologie , Cycle oestral/physiologie , ARN ribosomique 16S/génétique , Bactéries/classification , Bactéries/génétique , Bactéries/isolement et purification , Voies urinaires/microbiologie , Urine/microbiologie , ADN bactérien/génétique
15.
Physiol Behav ; 284: 114645, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39047942

RÉSUMÉ

Since the NIH 'Sex as biological variable' policy, the percentage of studies including female subjects have increased largely. Nonetheless, many researchers fail to adequate their protocols to include females. In this narrative review, we aim to discuss the methodological pitfalls of the inclusion of female rodents in behavioral neuroscience. We address three points to consider in studies: the manipulations conducted only in female animals (such as estrous cycle monitoring, ovariectomy, and hormone replacement), the consideration of males as the standard, and biases related to interpretation and publication of the results. In addition, we suggest guidelines and perspectives for the inclusion of females in preclinical research.


Sujet(s)
Neurosciences , Animaux , Femelle , Neurosciences/méthodes , Rodentia , Comportement animal/physiologie , Cycle oestral/physiologie , Caractères sexuels , Mâle
16.
Reprod Biol ; 24(3): 100918, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38924877

RÉSUMÉ

Apelin and APJ have been shown to regulate female reproductive functions. However, its uterine expression during the oestrous cycle and its regulation by ovarian steroids, along with gonadotropin regulation in the ovary, has not been investigated. This study aimed to analyze the steroid-dependent uterine expression of apelin/APJ in the uterus along with the oestrous cycle. Furthermore, it also aimed to investigate gonadotropin-dependent ovarian expression of apelin and APJ. To investigate the uterine expression of apelin and APJ during estrous cycle in mice, uterus at different estrous stage were collected. To explore the ovarian steroids dependent expression of apelin system in the uterus, ovariectomized mice were treated with only estrogen at dose of 30 ng/g, only progesterone at dose of 150 µg/g and combined doses. To study the effect of gonadotropin on ovarian expression of apelin system, immature mice were injected with 2.5 IU of pregnant mare serum gonadotropin (PMSG) alone and both PMSG plus 2.5 IU of chorionic gonadotropin (hCG). Apelin and APJ protein expression are modulated by estrous phases in the uterus. The uterine apelin and APJ expression are up-regulated by estrogen and down-regulated by progesterone. The expression and localization of APJ showed increased abundance in the follicles of PMSG treated mice, however, the PMSG plus HCG treatment showed formation of corpus luteum with increased abundance of APJ and progesterone secretion. The expression of apelin and APJ are regulated by pituitary gonadotropin in the ovary and uterine apelin system by ovarian steroid hormone.


Sujet(s)
Récepteur de l'apeline , Apeline , Ovaire , Progestérone , Utérus , Animaux , Femelle , Souris , Apeline/métabolisme , Récepteur de l'apeline/métabolisme , Gonadotrophine chorionique/pharmacologie , Oestrogènes/pharmacologie , Oestrogènes/métabolisme , Cycle oestral/métabolisme , Cycle oestral/physiologie , Gonadotrophine équine/pharmacologie , Ovariectomie , Ovaire/métabolisme , Ovaire/effets des médicaments et des substances chimiques , Progestérone/pharmacologie , Progestérone/métabolisme , Utérus/métabolisme , Utérus/effets des médicaments et des substances chimiques
17.
Reprod Sci ; 31(10): 3215-3227, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-38937400

RÉSUMÉ

To date, there is no comparative data on the effects of carbohydrates, fat, and proteins, which are macronutrients, on female reproductive functions. Therefore, in this study, we investigated the effects of diets enriched with carbohydrates, fats, and proteins on folliculogenesis and oocyte development in female rats. 21-day-old rats that were just weaned were divided into 4 groups: control, carbohydrate, fat, and protein. The control group was fed with standard chow and the carbohydrate, fat, and protein groups were fed diets enriched with 75% carbohydrate, 60% fat, and 50% protein for 11 weeks, respectively. It was found that high-fat and high-protein diets caused an increase in the estrous cycle length compared to carbohydrate group (p < 0.05). Graafian follicle number decreased in the protein group compared to the control (p < 0.05). However, the atretic follicle number was higher in the fat group compared to the control group (p < 0.05). In the carbohydrate group, Zp1 was found to be lower than the control and protein groups, Zp2 was found to be lower than the control, and Zp3 was found to be lower than the fat group (p < 0.05). While BMP15 was similar between groups (p > 0.05), GDF9 was lower in all diet groups compared to the control (p < 0.05). Foxo3a was lower in the protein group compared to carbohydrate and control (p < 0.05). GAS2 was found to be higher in the control group than the fat group, and higher in the carbohydrate group than the fat and protein groups (p < 0.05). FSH, LH, Progesterone, and E2 levels were higher in all three diet groups than in the control (p < 0.05). Also, significant differences were observed between the groups regarding adiponectin, resistin, and leptin levels. Taken together, high carbohydrate, fat, and protein intake are associated with impairment of the menstrual cycle, depletion of the developing follicle types, and altered expression of folliculogenesis-specific genes and hormones. Therefore, long-term macronutrient diets may result in shortened reproductive periods and reduced fertilization potential in females in the long run.


Sujet(s)
Hydrates de carbone alimentaires , Protéines alimentaires , Ovocytes , Follicule ovarique , Animaux , Femelle , Follicule ovarique/métabolisme , Follicule ovarique/effets des médicaments et des substances chimiques , Ovocytes/métabolisme , Protéines alimentaires/administration et posologie , Rats , Hydrates de carbone alimentaires/administration et posologie , Matières grasses alimentaires/administration et posologie , Ovogenèse/effets des médicaments et des substances chimiques , Ovogenèse/physiologie , Cycle oestral/physiologie , Alimentation riche en graisse , Rat Wistar , Régime riche en protéines
18.
Theriogenology ; 226: 228-235, 2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-38924892

RÉSUMÉ

Although anti-Müllerian hormone (AMH) is involved in the regulation of granulosa cell function in female animals, its role in tissues other than ovarian follicles remains poorly understood. It has also been suggested that cows with high circulating AMH concentrations have increased fertility; however, the mechanism has not been elucidated. This study was conducted to identify the presence of the AMH-signaling system and its target cells in the bovine corpus luteum formed from an ovulated follicle. Immunoblotting revealed that the proteolytically cleaved C-terminal region in AMH (AMHC), a biologically active peptide, was present in trace amounts in the early corpus luteum and significantly increased during the mid to regressed stages. AMHC and cleaved N-terminal region (AMHN) in AMH generate a noncovalent isoform that improves the activity of AMH signaling. An immunohistochemical analysis revealed that AMHC, AMHN, and type II AMH receptor (AMHR2) were localized to luteal cells during the entire estrous cycle. AMH in the corpus luteum seemed to be newly synthesized since AMH expression was detected. These findings suggest that AMH signaling is involved in the regulation of luteal cell function through an autocrine and post-translational processing mechanism. The level of AMHR2 and mRNA expression of AMHR2 and type I AMH receptors (activin-like kinase 2, 3, and 6) were highest in the mid stage. Thus, AMH signaling in the corpus luteum may also be regulated by changes in the receptor levels. Since the transforming growth factor-beta superfamily, to which AMH belongs, is a multifunctional polypeptide growth factor, further studies are needed to evaluate whether AMH signaling has a role in facilitating or inhibiting luteal cell functions.


Sujet(s)
Hormone antimullérienne , Corps jaune , Récepteurs peptidiques , Récepteurs TGF-bêta , Animaux , Femelle , Hormone antimullérienne/métabolisme , Hormone antimullérienne/génétique , Corps jaune/métabolisme , Bovins , Récepteurs peptidiques/métabolisme , Récepteurs peptidiques/génétique , Récepteurs TGF-bêta/métabolisme , Récepteurs TGF-bêta/génétique , Régulation de l'expression des gènes/physiologie , Cycle oestral/métabolisme , Cycle oestral/physiologie , ARN messager/métabolisme , ARN messager/génétique
19.
Neurosci Lett ; 836: 137888, 2024 Jul 27.
Article de Anglais | MEDLINE | ID: mdl-38945352

RÉSUMÉ

There are currently no FDA-approved treatments for cocaine use disorder. Recent preclinical and clinical studies showed that deep brain stimulation (DBS) in limbic regions reduced drug seeking behavior. Our previous work indicated that DBS of the nucleus accumbens shell attenuated reinstatement of cocaine seeking, a model of relapse, in male rats. The current experiments were designed to evaluate the effect of electrical DBS on cocaine reinstatement in female rats across the estrous cycle. Rats were allowed to self-administer cocaine and lever responding was subsequently extinguished. Cocaine seeking was reinstated by an acute injection of experimenter-delivered cocaine. The effect of nucleus accumbens shell DBS vs. sham stimulation on cocaine-primed reinstatement was evaluated in female and male rats using a within-subjects counterbalanced design. Consistent with previous work, accumbens shell DBS suppressed cocaine seeking in male rats. In sharp contrast, accumbens shell DBS had no effect on cocaine reinstatement in female rats evaluated in either the estrus or non-estrus phases. These results suggest that changes across the estrous cycle are not responsible for the differences in the effect of DBS on cocaine reinstatement between female and male rats.


Sujet(s)
Cocaïne , Stimulation cérébrale profonde , Comportement de recherche de substances , Cycle oestral , Noyau accumbens , Autoadministration , Animaux , Femelle , Mâle , Stimulation cérébrale profonde/méthodes , Rats , Noyau accumbens/effets des médicaments et des substances chimiques , Cocaïne/administration et posologie , Comportement de recherche de substances/physiologie , Comportement de recherche de substances/effets des médicaments et des substances chimiques , Cycle oestral/physiologie , Troubles liés à la cocaïne/thérapie , Troubles liés à la cocaïne/psychologie , Rat Sprague-Dawley , Extinction (psychologie)/effets des médicaments et des substances chimiques , Caractères sexuels
20.
Horm Behav ; 164: 105593, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38909429

RÉSUMÉ

Autism Spectrum Disorder (ASD) is characterized by differences in social communication and interaction, as well as areas of focused interests and/or repetitive behaviors. Recent studies have highlighted a higher prevalence of endocrine and reproductive disturbances among females on the autism spectrum, hinting at potential disruptions within the hypothalamus-pituitary-ovary (HPO) axis. This research aims to explore the reproductive health disparities in ASD using an animal model of autism, the C58/J inbred mouse strain, with a focus on reproductive performance and hormonal profiles compared to the C57BL/6J control strain. Our findings revealed that the estrous cycle in C58/J females is disrupted, as evidenced by a lower frequency of complete cycles and a lack of cyclical release of estradiol and progesterone compared to control mice. C58/J females also exhibited poor performance in several reproductive parameters, including reproductive lifespan and fertility index. Furthermore, estrogen receptor alpha content showed a marked decrease in the hypothalamus of C58/J mice. These alterations in the estrous cycle, hormonal imbalances, and reduced reproductive function imply dysregulation in the HPO axis. Additionally, our in-silico study identified a group of genes involved in infertility carrying single-nucleotide polymorphisms (SNPs) in the C58/J strain, which also have human orthologs associated with autism. These findings could offer valuable insights into the molecular underpinnings of neuroendocrine axis disruption and reproductive issues observed in ASD.


Sujet(s)
Modèles animaux de maladie humaine , Hypothalamus , Souris de lignée C57BL , Animaux , Femelle , Souris , Hypothalamus/métabolisme , Cycle oestral/physiologie , Santé reproductive , Trouble autistique/métabolisme , Trouble autistique/génétique , Trouble du spectre autistique/métabolisme , Trouble du spectre autistique/génétique , Reproduction/physiologie , Reproduction/génétique , Progestérone/sang , Progestérone/métabolisme , Oestradiol/sang , Oestradiol/métabolisme , Mâle , Hormones sexuelles stéroïdiennes/métabolisme , Hormones sexuelles stéroïdiennes/sang
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE