Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 308
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 185(4): 654-671.e22, 2022 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-35065713

RESUMEN

Sex hormones exert a profound influence on gendered behaviors. How individual sex hormone-responsive neuronal populations regulate diverse sex-typical behaviors is unclear. We performed orthogonal, genetically targeted sequencing of four estrogen receptor 1-expressing (Esr1+) populations and identified 1,415 genes expressed differentially between sexes or estrous states. Unique subsets of these genes were distributed across all 137 transcriptomically defined Esr1+ cell types, including estrous stage-specific ones, that comprise the four populations. We used differentially expressed genes labeling single Esr1+ cell types as entry points to functionally characterize two such cell types, BNSTprTac1/Esr1 and VMHvlCckar/Esr1. We observed that these two cell types, but not the other Esr1+ cell types in these populations, are essential for sex recognition in males and mating in females, respectively. Furthermore, VMHvlCckar/Esr1 cell type projections are distinct from those of other VMHvlEsr1 cell types. Together, projection and functional specialization of dimorphic cell types enables sex hormone-responsive populations to regulate diverse social behaviors.


Asunto(s)
Ciclo Estral/genética , Regulación de la Expresión Génica , Caracteres Sexuales , Conducta Sexual Animal/fisiología , Agresión , Animales , Aromatasa/metabolismo , Trastorno Autístico/genética , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neuronas/metabolismo , Conducta Social
2.
Nature ; 599(7885): 436-441, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34732894

RESUMEN

The state of somatic energy stores in metazoans is communicated to the brain, which regulates key aspects of behaviour, growth, nutrient partitioning and development1. The central melanocortin system acts through melanocortin 4 receptor (MC4R) to control appetite, food intake and energy expenditure2. Here we present evidence that MC3R regulates the timing of sexual maturation, the rate of linear growth and the accrual of lean mass, which are all energy-sensitive processes. We found that humans who carry loss-of-function mutations in MC3R, including a rare homozygote individual, have a later onset of puberty. Consistent with previous findings in mice, they also had reduced linear growth, lean mass and circulating levels of IGF1. Mice lacking Mc3r had delayed sexual maturation and an insensitivity of reproductive cycle length to nutritional perturbation. The expression of Mc3r is enriched in hypothalamic neurons that control reproduction and growth, and expression increases during postnatal development in a manner that is consistent with a role in the regulation of sexual maturation. These findings suggest a bifurcating model of nutrient sensing by the central melanocortin pathway with signalling through MC4R controlling the acquisition and retention of calories, whereas signalling through MC3R primarily regulates the disposition of calories into growth, lean mass and the timing of sexual maturation.


Asunto(s)
Desarrollo Infantil/fisiología , Estado Nutricional/fisiología , Pubertad/fisiología , Receptor de Melanocortina Tipo 3/metabolismo , Maduración Sexual/fisiología , Adolescente , Anciano de 80 o más Años , Animales , Niño , Ciclo Estral/genética , Ciclo Estral/fisiología , Femenino , Homocigoto , Humanos , Hipotálamo/citología , Hipotálamo/fisiología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Melanocortinas/metabolismo , Menarquia/genética , Menarquia/fisiología , Ratones , Fenotipo , Pubertad/genética , Receptor de Melanocortina Tipo 3/deficiencia , Receptor de Melanocortina Tipo 3/genética , Maduración Sexual/genética , Factores de Tiempo , Aumento de Peso
3.
J Mammary Gland Biol Neoplasia ; 29(1): 13, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38916673

RESUMEN

Conflicting data exist as to how mammary epithelial cell proliferation changes during the reproductive cycle. To study the effect of endogenous hormone fluctuations on gene expression in the mouse mammary gland, we performed bulk RNAseq analyses of epithelial and stromal cell populations that were isolated either during puberty or at different stages of the adult virgin estrous cycle. Our data confirm prior findings that proliferative changes do not occur in every mouse in every cycle. We also show that during the estrous cycle the main gene expression changes occur in adipocytes and fibroblasts. Finally, we present a comprehensive overview of the Wnt gene expression landscape in different mammary gland cell types in pubertal and adult mice. This work contributes to understanding the effects of physiological hormone fluctuations and locally produced signaling molecules on gene expression changes in the mammary gland during the reproductive cycle and should be a useful resource for future studies investigating gene expression patterns in different cell types across different developmental timepoints.


Asunto(s)
Células Epiteliales , Perfilación de la Expresión Génica , Glándulas Mamarias Animales , Maduración Sexual , Células del Estroma , Transcriptoma , Animales , Femenino , Ratones , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Células del Estroma/metabolismo , Células Epiteliales/metabolismo , Perfilación de la Expresión Génica/métodos , Maduración Sexual/fisiología , Proliferación Celular , Ciclo Estral/genética
4.
Mamm Genome ; 34(4): 559-571, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37656189

RESUMEN

The spontaneous mutation stubby (stb) in mice causes chondrodysplasia and male infertility due to impotence through autosomal recessive inheritance. In this study, we conducted linkage analysis to localize the stb locus within a 1.6 Mb region on mouse chromosome 2 and identified a nonsense mutation in Adamtsl2 of stb/stb mice. Histological analysis revealed disturbed endochondral ossification with a reduced hypertrophic chondrocyte layer and stiff skin with a thickened dermal layer. These phenotypes are similar to those observed in humans and mice with ADAMTSL2/Adamtsl2 mutations. Moreover, stb/stb female mice exhibited severe uterine hypoplasia at 5 weeks of age and irregular estrous cycles at 10 weeks of age. In normal mice, Adamtsl2 was more highly expressed in the ovary and pituitary gland than in the uterus, and this expression was decreased in stb/stb mice. These findings suggest that Adamtsl2 may function in these organs rather than in the uterus. Thus, we analyzed Gh expression in the pituitary gland and plasma estradiol and IGF1 levels, which are required for the development of the female reproductive tract. There was no significant difference in Gh expression and estradiol levels, whereas IGF1 levels in stb/stb mice were significantly reduced to 54-59% of those in +/+ mice. We conclude that Adamtsl2 is required for the development of the uterus and regulation of the estrous cycle in female mice, and decreased IGF1 may be related to these abnormalities.


Asunto(s)
Codón sin Sentido , Estradiol , Humanos , Animales , Ratones , Masculino , Femenino , Codón sin Sentido/genética , Mutación , Útero , Ciclo Estral/genética , Proteínas ADAMTS/genética , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo
5.
Horm Behav ; 156: 105439, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37813043

RESUMEN

Anxiety disorders are the most prevalent mental illnesses worldwide, exhibit high heritability, and affect twice as many women as men. To evaluate potential interactions between genetic background and cycling ovarian hormones on sex differences in susceptibility to negative valence behaviors relevant to anxiety disorders, we assayed avoidance behavior and cued threat memory dynamics in gonadally-intact adult male and female mice across four common inbred mouse strains: C57Bl/6J, 129S1/SVlmJ, DBA/2J, and BALB/cJ. Independent of sex, C57Bl/6J mice exhibited low avoidance but high threat memory, 129S1/SvlmJ mice high avoidance and high threat memory, DBA/2J mice low avoidance and low threat memory, and BALB/cJ mice high avoidance but low threat memory. Within-strain comparisons revealed reduced avoidance behavior in the high hormone phase of the estrous cycle (proestrus) compared to all other estrous phases in all strains except DBA/2J, which did not exhibit cycle-dependent behavioral fluctuations. Robust and opposing sex differences in threat conditioning and extinction training were found in the C57Bl/6J and 129S1/SvlmJ lines, whereas no sex differences were observed in the DBA/2J or BALB/cJ lines. C57Bl/6J males exhibited enhanced acute threat memory, whereas 129S1/SvlmJ females exhibited enhanced sustained threat memory, compared to their sex-matched littermates. These effects were not mediated by estrous cycle stage or sex differences in active versus passive defensive behavioral responses. Our data demonstrate that core features of behavioral endophenotypes relevant to anxiety disorders, such as avoidance and threat memory, are genetically driven yet dissociable and can be influenced further by cycling ovarian hormones.


Asunto(s)
Reacción de Prevención , Conducta Animal , Humanos , Ratones , Femenino , Masculino , Animales , Ratones Endogámicos DBA , Conducta Animal/fisiología , Reacción de Prevención/fisiología , Caracteres Sexuales , Ciclo Estral/genética , Ratones Endogámicos C57BL , Antecedentes Genéticos , Hormonas , Especificidad de la Especie
6.
Anim Biotechnol ; 34(9): 4569-4579, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36752221

RESUMEN

Maiwa yak is a special breed of animal living on the Qinghai-Tibet Plateau, which has great economic value, but its fertility rate is low. The corpus luteum (CL) is a temporary tissue that plays a crucial role in maintaining the physiological cycle. However, little is known about the transcriptome profile in Maiwa yak CL. In the present study, the transcriptome of Maiwa yak CL at early (EYCL), middle (MYCL) and late-stages (LYCL) was studied employing high-throughput sequencing. A total of 25,922 transcripts were identified, including 22,277 known as well as 3,645 novel ones. Furthermore, 690 and 212 differentially expressed (DE) mRNAs were detected in the EYCL vs. MYCL and MYCL vs. LYCL groups, respectively. KEGG pathway enrichment analysis of DEGs illustrated that the most enriched pathway was PI3K-Akt pathway. Furthermore, twenty-six DEGs were totally found to be associated with different biological processes of CL development. One of these genes, PGRMC1, displayed a dynamical expression trend during the lifespan of yak CL. The knockdown of PGRMC1 in luteinized yak granulosa cells resulted in defective steroidogenesis. In conclusion, this study analyzed the transcriptome profiles in yak CL of different stages, and provided a novel database for analyzing the gene network in yak CL.HIGHLIGHTSThe manuscript analyzed the transcriptome profiles in yak CL during the estrous cycle.Twenty-six DEGs were found to be associated with the development or function of CL.One of the DEGs, PGRMC1, was found to be responsible for steroidogenesis in luteinized yak granulosa cells.


Asunto(s)
Perfilación de la Expresión Génica , Fosfatidilinositol 3-Quinasas , Femenino , Bovinos/genética , Animales , Perfilación de la Expresión Génica/veterinaria , Cuerpo Lúteo , Ciclo Estral/genética , Transcriptoma/genética
7.
Anim Biotechnol ; 34(7): 2554-2564, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35913775

RESUMEN

Estrus detection is a major problem in buffaloes because of the poor expression of estrus signs leading to low reproductive efficiency. Salivary transcripts analysis is a promising tool to identify biomarkers; therefore, the present study was carried out to evaluate their potential as estrus biomarkers. The levels of HSD17B1, INHBA, HSPA1A, TES transcripts were compared in saliva during estrous cycle stages [early proestrus (day -2, EP), late proestrus (day-1, LP), estrus (E), metestrus (ME) and diestrus (DE)] of cyclic heifers (n = 8) and pluriparous (n = 8) buffaloes by employing quantitative real-time polymerase chain reaction (qRT-PCR). The levels of HSD17B1 (EP/DE 1.46-2.43 fold, LP/DE 2.49-3.06 fold; E/DE 7.21-11.9-fold p < 0.01; ME/D 1.0-1.16 fold) and HSPA1A (EP/DE 0.93-2.39 fold, LP/DE 2.68-3.23 fold; E/DE 8.52-15.18 fold p < 0.01; ME/D 0.86-1.01 fold) were significantly altered during the estrus than other estrous cycle stages in both cyclic heifers and pluriparous buffaloes. Receiver operating characteristic curve analysis revealed the ability of salivary HSD17B1 (AUC 0.96; p < 0.001) and HSPA1A (AUC 0.99; p < 0.01) to differentiate E from other stages of the estrous cycle. Significantly higher levels of HSD17B1 and HSPA1A transcripts in saliva during the estrus phase suggest their biomarkers potential for estrus detection in buffaloes.


Asunto(s)
Búfalos , Estro , Femenino , Animales , Bovinos/genética , Búfalos/genética , Ciclo Estral/genética , Biomarcadores
8.
Dev Biol ; 476: 240-248, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33864778

RESUMEN

Female fertility in mammals requires iterative remodeling of the entire adult female reproductive tract across the menstrual/estrous cycle. However, while transcriptome dynamics across the estrous cycle have been reported in human and bovine models, no global analysis of gene expression across the estrous cycle has yet been reported for the mouse. Here, we examined the cellular composition and global transcriptional dynamics of the mouse oviduct along the anteroposterior axis and across the estrous cycle. We observed robust patterns of differential gene expression along the anteroposterior axis, but we found surprisingly few changes in gene expression across the estrous cycle. Notable gene expression differences along the anteroposterior axis included a surprising enrichment for genes related to embryonic development, such as Hox and Wnt genes. The relatively stable transcriptional dynamics across the estrous cycle differ markedly from other mammals, leading us to speculate that this is an evolutionarily derived state that may reflect the extremely rapid five-day mouse estrous cycle. This dataset fills a critical gap by providing an important genomic resource for a highly tractable genetic model of mammalian female reproduction.


Asunto(s)
Fertilidad/genética , Oviductos/metabolismo , Transcriptoma/genética , Animales , Desarrollo Embrionario/genética , Ciclo Estral/genética , Femenino , Fertilidad/fisiología , Expresión Génica/genética , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/genética , Ratones , Oviductos/fisiología , Embarazo
9.
FASEB J ; 35(12): e22000, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34731497

RESUMEN

Extracellular vesicles (EVs) found in various biological fluids and particularly in reproductive fluids, have gained considerable attention for their possible role in cell- to- cell communication. Among, the different bioactive molecules cargos of EVs, MicroRNAs (miRNAs) are emerging as promising diagnostic biomarkers with high clinical potential. Aiming to understand the roles of EVs in bovine reproductive tract, we intended to characterize and profile the EVs of oviduct and uterine fluids (OF-EVs, UF-EVs) and their miRNA across the estrous cycle. Nanoparticle tracking analysis and transmission electron microscopy confirmed the existence of small EV population in OF and UF at all stages, (size between 30 and 200 nm; concentration: 3.4 × 1010  EVs/ml and 6.0 × 1010  EVs/ml for OF and UF, respectively, regardless of stage). The identification of EV markers (CD9, HSP70, and ALIX proteins) was confirmed by western blot. The miRNA analysis revealed the abundance of 310 and 351 miRNAs in OF-EVs and UF-EVs, respectively. Nine miRNAs were differentially abundant in OF-EVs between stages of the cycle, eight of them displayed a progressive increase from S1 to S4 (p < .05). In UF-EVs, a total of 14 miRNAs were differentially abundant between stages. Greater differences were observed between stage 1 (S1) and stage 3 (S3), with 11 miRNAs enriched in S3 compared to S1. Functional enrichment analysis revealed the involvement of these miRNAs in relevant pathways such as cell signaling, intercellular junctions, and reproductive functions that may be implicated in oviduct and uterus modulation across the cycle, but also in their preparation for embryo/conceptus presence and development.


Asunto(s)
Comunicación Celular , Ciclo Estral/metabolismo , Vesículas Extracelulares/metabolismo , MicroARNs/genética , Oviductos/metabolismo , Útero/metabolismo , Animales , Bovinos , Ciclo Estral/genética , Vesículas Extracelulares/genética , Femenino , MicroARNs/metabolismo , Fagocitosis
10.
J Dairy Sci ; 105(2): 1674-1686, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34799112

RESUMEN

This observational study aimed to determine the effect of genetic merit for fertility traits on estrous expression and estrous cycle duration in grazing dairy cows, as measured by an activity monitoring device. A secondary aim was to describe changes in expression of estrus that occur during successive estrous cycles postpartum. Neck-mounted, activity-monitoring devices (Heatime, SCR Engineers Ltd.) were fitted to nulliparous Holstein-Friesian heifers with positive (POS FertBV) or negative genetic merit for fertility traits (NEG FertBV) to capture activity data during their first and second lactations (POS FertBV: n = 242, n = 188; NEG FertBV: n = 159, n = 87 in lactation 1 and 2, respectively). An estrous event was identified when the activity change index exceeded 26 activity units (AU) for 4 h. A total of 1,254 and 892 estrous events were identified in lactation 1 and 2, respectively. Estrous duration was defined as the interval between when the threshold was first exceeded and when activity dropped below the threshold, with no new event starting within 24 h of the end of the previous event. This definition of estrus included cows in which activity crossed the threshold multiple times in a day and were classified as a single estrous event. A second measure, high activity duration, was defined as the total hours that activity exceeded the threshold. To characterize estrous activity, peak activity (above baseline) and total activity (area under the curve of activity above baseline) were measured. Compared with NEG FertBV cows, POS FertBV cows had more active, longer estrous events. In lactation 1, the POS FertBV group had a mean estrous duration and a high activity duration of 12.5 and 12.4 h compared with 11.4 and 11.3 h for the NEG FertBV group [standard error of the difference (SED) = 0.5 and 0.4 h, respectively]. This significant difference also occurred in lactation 2, with a mean estrous duration of 13.1 versus 11.8 h (SED = 0.5 h) and a high activity duration of 13.0 versus 11.8 h (SED = 0.4 h) in the POS and NEG FertBV groups, respectively. Total activity and peak activity were greater in the POS compared with the NEG FertBV group in lactation 1 (peak activity: 65.5 vs. 55.8 AU, SED = 2.4 AU; total activity: 588 vs. 494 AU, SED = 25 AU) and lactation 2 (peak activity: 72.5 vs. 61.2 AU, SED = 2.9 AU; total activity: 648 vs. 541 AU, SED = 30 AU). Estrous cycle duration did not differ between the POS and NEG FertBV groups (lactation 1: 20.4 vs. 20.6 d, SED = 0.25; lactation 2: 20.8 vs. 21.0 d, SED = 0.28). Less estrous activity of the cow was associated with the first postpartum estrus. In contrast, the number of previous estrous events did not consistently affect the duration of the subsequent estrous cycle. The outcomes of this study provide evidence that positive genetic merit for fertility traits is associated with more overt estrous expression. Selection for these traits may improve estrous expression and thus estrous detection in commercial herds.


Asunto(s)
Estro , Lactancia , Animales , Bovinos/genética , Ciclo Estral/genética , Estro/genética , Femenino , Fertilidad/genética , Fenotipo , Progesterona
11.
Genomics ; 113(1 Pt 1): 366-379, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33309770

RESUMEN

To better understand the molecular basis of corpus luteum (CL) development and function RNA-Seq was utilized to identify differentially expressed genes (DEGs) in porcine CL during different physiological stages of the estrous cycle viz. early (EL), mid (ML), late (LL) and regressed (R) luteal. Stage wise comparisons obtained 717 (EL vs. ML), 568 (EL vs. LL), 527 (EL vs. R), 786 (ML vs. LL), 474 (ML vs. R) and 534 (LL vs. R) DEGs with log2(FC) ≥1 and p < 0.05. The process of angiogenesis, steroidogenesis, signal transduction, translation, cell proliferation and tissue remodelling were significantly (p < 0.05) enriched in EL, ML and LL stages, where as apoptosis was most active in regressed stage. Pathway analysis revealed that most annotated genes were associated with lipid metabolism, translation, immune and endocrine system pathways depicting intra-luteal control of diverse CL function. The network analysis identified genes AR, FOS, CDKN1A, which were likely the novel hub genes regulating CL physiology.


Asunto(s)
Cuerpo Lúteo/crecimiento & desarrollo , Ciclo Estral/genética , Porcinos/genética , Transcriptoma , Animales , Cuerpo Lúteo/metabolismo , Ciclo Estral/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Redes y Vías Metabólicas/genética , Porcinos/crecimiento & desarrollo , Porcinos/fisiología
12.
Int J Mol Sci ; 23(13)2022 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-35806363

RESUMEN

Endometrosis is a frequently occurring disease decreasing mares' fertility. Thus, it is an important disease of the endometrium associated with epithelial and stromal cell alterations, endometrium gland degeneration and periglandular fibrosis. Multiple degenerative changes are found in uterine mucosa, the endometrium. However, their pathogenesis is not well known. It is thought that nuclear factor-κB (NF-κB), a cell metabolism regulator, and its activation pathways take part in it. The transcription of the profibrotic pathway genes of the NF-κB in fibrotic endometria differed between the follicular (FLP) and mid-luteal (MLP) phases of the estrous cycle, as well as with fibrosis progression. This study aimed to investigate the transcription of genes of estrogen (ESR1, ESR2) and progesterone receptors (PGR) in equine endometria to find relationships between the endocrine environment, NF-κB-pathway, and fibrosis. Endometrial samples (n = 100), collected in FLP or MLP, were classified histologically, and examined using quantitative PCR. The phase of the cycle was determined through the evaluation of ovarian structures and hormone levels (estradiol, progesterone) in serum. The transcription of ESR1, ESR2, and PGR decreased with the severity of endometrial fibrosis and degeneration of the endometrium. Moreover, differences in the transcription of ESR1, ESR2, and PGR were noted between FLP and MLP in the specific categories and histopathological type of equine endometrosis. In FLP and MLP, specific moderate and strong correlations between ESR1, ESR2, PGR and genes of the NF-κB pathway were evidenced. The transcription of endometrial steroid receptors can be subjected to dysregulation with the degree of equine endometrosis, especially in both destructive types of endometrosis, and mediated by the canonical NF-κB pathway depending on the estrous cycle phase.


Asunto(s)
Enfermedades del Ovario , Receptores de Esteroides , Animales , Endometrio/metabolismo , Ciclo Estral/genética , Femenino , Fibrosis , Caballos , Humanos , FN-kappa B/genética , FN-kappa B/metabolismo , Enfermedades del Ovario/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo
13.
Dev Biol ; 458(1): 43-51, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31610144

RESUMEN

The steroid hormones are instrumental for the growth of mammary epithelial cells. Our previous study indicates that hormones regulate the expression of Rspondin-1 (Rspo1). Yet, the regulatory mechanism remains unknown. In the current study, we identify Amphiregulin (Areg) as a novel upstream regulator of Rspo1 expression mediating the hormonal influence. In response to hormonal signaling, Areg emanating from estrogen receptor (ER)-positive luminal cells, induce the expression of Rspo1 in ER-negative luminal cells. The paracrine action of Areg on Rspo1 expression is dependent on Egfr. Our data reveal a novel Estrogen-Areg-Rspo1 regulatory axis in the mammary gland, providing new evidence for the orchestrated action of systemic hormones and local growth factors.


Asunto(s)
Anfirregulina/fisiología , Estradiol/fisiología , Ciclo Estral/fisiología , Regulación de la Expresión Génica/fisiología , Glándulas Mamarias Animales/metabolismo , Progesterona/fisiología , Trombospondinas/biosíntesis , Anfirregulina/genética , Animales , Células Cultivadas , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/fisiología , Clorhidrato de Erlotinib/farmacología , Estradiol/farmacología , Ciclo Estral/genética , Femenino , Glándulas Mamarias Animales/citología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Cultivo Primario de Células , Progesterona/farmacología , ARN Interferente Pequeño/genética , Trombospondinas/genética , Transcriptoma
14.
Am J Physiol Endocrinol Metab ; 320(3): E566-E580, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33427045

RESUMEN

Sex as a biological variable has been the focus of increasing interest. Relatively few studies have focused, however, on differences in peripheral taste function between males and females. Nonetheless, there are reports of sex-dependent differences in chemosensitivity in the gustatory system. The involvement of endogenous changes in ovarian hormones has been suggested to account for taste discrepancies. Additionally, whether sex differences exist in taste receptor expression, activation, and subsequent signaling pathways that may contribute to different taste responsiveness is not well understood. In this study, we show the presence of both the nuclear and plasma membrane forms of estrogen receptor (ER) mRNA and protein in mouse taste cells. Furthermore, we provide evidence that estrogen increases taste cell activation during the application of fatty acids, the chemical cue for fat taste, in taste receptor cells. We found that genes important for the transduction pathway of fatty acids vary between males and females and that these differences also exist across the various taste papillae. In vivo support for the effect of estrogens in taste cells was provided by comparing the fatty acid responsiveness in male, intact female, and ovariectomized (OVX) female mice with and without hormone replacement. In general, females detected fatty acids at lower concentrations, and the presence of circulating estrogens increased this apparent fat taste sensitivity. Taken together, these data indicate that increased circulating estrogens in the taste system may play a significant role in physiology and chemosensory cellular activation and, in turn, may alter taste-driven behavior.NEW & NOTEWORTHY Using molecular, cellular, and behavioral analyses, this study shows that sex differences occur in fat taste in a mouse model. Female mice are more responsive to fatty acids, leading to an overall decrease in intake and fatty acid preference. These differences are linked to sex hormones, as estradiol enhances taste cell responsiveness to fatty acids during periods of low circulating estrogen following ovariectomy and in males. Estradiol is ineffective in altering fatty acid signaling during a high-estrogen period and in ovariectomized mice on hormone replacement. Thus, taste receptor cells are a direct target for actions of estrogen, and there are multiple receptors with differing patterns of expression in taste cells.


Asunto(s)
Grasas de la Dieta/farmacología , Estradiol/sangre , Papilas Gustativas/efectos de los fármacos , Gusto/fisiología , Animales , Células Cultivadas , Grasas de la Dieta/metabolismo , Ciclo Estral/genética , Ciclo Estral/metabolismo , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ovariectomía , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Caracteres Sexuales , Gusto/efectos de los fármacos , Papilas Gustativas/metabolismo , Percepción del Gusto/fisiología
15.
Biol Reprod ; 105(5): 1205-1220, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34333627

RESUMEN

Members of the differential screening-selected gene aberrative in neuroblastoma (DAN) protein family are developmentally conserved extracellular binding proteins that antagonize bone morphogenetic protein (BMP) signaling. This protein family includes the Gremlin proteins, GREM1 and GREM2, which have key functions during embryogenesis and adult physiology. While BMPs play essential roles in ovarian follicle development, the role of the DAN family in female reproductive physiology is less understood. We generated mice null for Grem2 to determine its role in female reproduction in addition to screening patients with primary ovarian insufficiency (POI) for variants in GREM2. Grem2-/- mice are viable, but female Grem2-/- mice have diminished fecundity and irregular estrous cycles. This is accompanied by significantly reduced production of ovarian anti-Müllerian hormone (AMH) from small growing follicles, leading to a significant decrease in serum AMH. Surprisingly, as AMH is a well-established marker of the ovarian reserve, morphometric analysis of ovarian follicles showed maintenance of primordial follicles in Grem2-/- mice like wild-type (WT) littermates. While Grem2 mRNA transcripts were not detected in the pituitary, Grem2 is expressed in hypothalami of WT female mice, suggesting the potential for dysfunction in multiple tissues composing the hypothalamic-pituitary-ovarian axis that contribute to the subfertility phenotype. Additionally, screening 106 women with POI identified one individual with a heterozygous variant in GREM2 that lies within the predicted BMP-GREM2 interface. In total, these data suggest that Grem2 is necessary for female fecundity by playing a novel role in regulating the HPO axis and contributing to female reproductive disease.


Asunto(s)
Citocinas/genética , Ciclo Estral/genética , Fertilidad/genética , Insuficiencia Ovárica Primaria/genética , Transducción de Señal , Animales , Citocinas/metabolismo , Femenino , Humanos , Ratones , Periodicidad
16.
J Reprod Dev ; 67(3): 217-221, 2021 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-33840679

RESUMEN

Functional relationship between nuclear receptor subfamily 4 group A member 3 (Nr4a3) and annexin A5 (Anxa5), which are two gonadotropin-releasing hormone (GnRH)-inducible genes, has been established while evaluating pituitary gonadotropes in relation to follicle-stimulating hormone beta (Fshb) expression. However, the physiological variations that arise due to the differential expression of these genes in the pituitary gland during rat estrous cycle remain unknown. This study aimed to evaluate the Nr4a3 and Anxa5 mRNA expression during the estrous cycle in rats in comparison with the expression of the gonadotropin subunit genes, luteinizing hormone beta (Lhb) and Fshb. Nr4a3 mRNA expression showed a single peak at 1400 h of proestrus during the 4-d estrous cycle. Anxa5 mRNA level was elevated along with increased Fshb mRNA expression after the decline of Nr4a3 mRNA until 2300 h. Lhb mRNA expression levels were not significantly changed during the estrous cycle. Notably, addition of a GnRH antagonist at 1100 h completely eradicated luteinizing hormone secretion at 1400 h and 1700 h of proestrus, and significantly reduced the Nr4a3 mRNA expression level at both the time points. These results suggest that GnRH is, at least partly, responsible for the increase in pituitary Nr4a3, and that the interaction between NR4A3 and ANXA5 is required to regulate Fshb expression during the preovulatory gonadotropin surge.


Asunto(s)
Anexina A5/metabolismo , Proteínas de Unión al ADN/metabolismo , Ciclo Estral/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Hipófisis/metabolismo , Animales , Anexina A5/genética , Proteínas de Unión al ADN/genética , Ciclo Estral/genética , Femenino , Regulación de la Expresión Génica , Hormona Liberadora de Gonadotropina/análogos & derivados , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Hormona Liberadora de Gonadotropina/farmacología , Antagonistas de Hormonas/farmacología , Hormona Luteinizante/sangre , Proteínas del Tejido Nervioso/genética , Hipófisis/efectos de los fármacos , Ratas , Receptores de HFE/genética , Receptores de HFE/metabolismo
17.
Reprod Domest Anim ; 56(3): 427-436, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33314336

RESUMEN

Follistatin-like 3 (FSTL3) is a regulator of cellular apoptosis and was previously identified via RNA-Seq to be associated with follicular development in mammalian ovaries. However, the mechanism underlying the FSTL3 regulation of oestrus in sheep remained poorly understood. In this study, the oestrogen (E2) and progesterone (P4) concentrations in blood were detected, and the expression level and functional analysis of FSTL3 in the ovary were studied during the different reproductive stage in Aohan fine wool sheep (seasonal breeding breed in China). The concentrations of E2 and P4 at the anestrus were significantly lower compared to dioestrus, proestrus and oestrus stages. Higher expression levels of FSTL3 were observed in the sheep ovary, hypothalamus, and thyroid. During different reproductive stages, higher expression levels were found during the stages of dioestrus and proestrus, while lower levels were found during the oestrus and anestrus stages. Functional analysis of FSTL3 was performed in primary granulosa cells (GCs) of sheep. The concentration of E2 increased significantly after RNAi interference of FSTL3, while the P4 level decreased. FSTL3 can decrease P4 levels, which might be involved in mediating oestrous cycle in sheep.


Asunto(s)
Ciclo Estral/metabolismo , Ovario/metabolismo , Ovinos/genética , Animales , Estrógenos/sangre , Ciclo Estral/genética , Femenino , Proteínas Relacionadas con la Folistatina/genética , Proteínas Relacionadas con la Folistatina/metabolismo , Expresión Génica , Progesterona/sangre , Ovinos/metabolismo
18.
Reprod Domest Anim ; 56(1): 46-57, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33098173

RESUMEN

To investigate the regulatory mechanism of the follicular-luteal phase transition in Turpan black sheep (Ovis aries), the genome-wide expression patterns of microRNAs (miRNAs) and genes were investigated in ovaries of six sheep (3 years and single lamb with 3 consecutive births) during follicular and luteal phases of the oestrous cycle. Bioinformatic analysis was used to screen potential miRNAs and genes related to Turpan black sheep ovarian function. RT-qPCR was used to validate the sequencing results. In total, we identified 139 known and 71 novel miRNAs in the two phases with miRNA-seq, and a total of 19 miRNAs were significantly differentially expressed, of which 7 were up-regulated and 12 were down-regulated in the follicular phase compared with luteal phase. A total of 150 genes were significantly differentially expressed, including 63 up-regulated and 87 down-regulated in the follicular phase compared with the luteal phase by RNA-seq data analysis. Those DEGs were significantly enriched in 103 GO terms and several KEGG pathways, including metabolic pathway, ovarian steroidogenesis, steroid hormone biosynthesis and oestrogen signalling pathway. In addition, we created a miRNA-mRNA regulatory network to further elucidate the mechanism of follicular-luteal transition. Finally, we identified key miRNAs and genes including miR-143, miR-99a, miR-150, miR-27a, miR-125b, STAR, STAT1, which might play crucial roles in reproductive hormone biosynthesis and follicular development. The miRNA-mRNA interactive network clearly illustrates molecular basis involving in follicular-luteal transition.


Asunto(s)
Ciclo Estral/genética , Ciclo Estral/fisiología , MicroARNs/metabolismo , ARN Mensajero/metabolismo , Oveja Doméstica , Animales , Femenino , Fase Folicular , Perfilación de la Expresión Génica/veterinaria , Fase Luteínica , Ovario/metabolismo
19.
Int J Mol Sci ; 22(5)2021 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-33800928

RESUMEN

Long non-coding RNAs (lncRNAs) play important roles in multiple biological processes including ovarian follicular development. Here we aimed to gain novel information regarding lncRNAs transcriptome profiles in porcine granulosa cells of advanced atretic antral (AA) and healthy antral (HA) follicles using RNA-seq. A total of 11,321 lncRNAs including 10,813 novel and 508 annotated lncRNAs were identified, of which 173 lncRNAs were differentially expressed (DE-lncRNAs); ten of these were confirmed by qRT-PCR. Gene Ontology indicated that DE-lncRNAs associated with developmental processes were highly enriched. Pathway analysis demonstrated predicted cis- and trans-targets of DE-lncRNAs. Potential mRNA targets of up-regulated DE-lncRNAs were mainly enriched in apoptosis related pathways, while targeted genes of downregulated DE-lncRNAs were primarily enriched in metabolism and ovarian steroidogenesis pathways. Linear regression analyses showed that expression of upregulated DE-lncRNAs was significantly associated with apoptosis related genes. NOVEL_00001850 is the most-downregulated DE-lncRNA (FDR = 0.04, FC = -6.53), of which miRNA binding sites were predicted. KEGG analysis of its downregulated target genes revealed that ovarian steroidogenesis was the second most highlighted pathway. qRT-PCR and linear regression analysis confirmed the expression and correlation of its potential targeted gene, CYP19A1, a key gene involved in estradiol synthesis. Our results indicate that lncRNAs may participate in granulosa cells apoptosis and thus antral follicular atresia.


Asunto(s)
Apoptosis/genética , Células de la Granulosa/metabolismo , Folículo Ovárico/metabolismo , ARN Largo no Codificante/genética , Animales , Ciclo Estral/genética , Femenino , Regulación de la Expresión Génica , Ontología de Genes , Modelos Lineales , MicroARNs/genética , MicroARNs/metabolismo , ARN Circular/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Porcinos , Transcriptoma , Regulación hacia Arriba
20.
Int J Mol Sci ; 22(2)2021 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-33477993

RESUMEN

Information on molecular mechanisms through which sex-steroids regulate oviductal function to support early embryo development is lacking. Here, we hypothesized that the periovulatory endocrine milieu affects the miRNA processing machinery and miRNA expression in bovine oviductal tissues. Growth of the preovulatory follicle was controlled to obtain cows that ovulated a small follicle (SF) and subsequently bore a small corpus luteum (CL; SF-SCL) or a large follicle (LF) and large CL (LF-LCL). These groups differed in the periovulatory plasmatic sex-steroid's concentrations. Ampulla and isthmus samples were collected on day four of the estrous cycle. Abundance of DROSHA, DICER1, and AGO4 transcripts was greater in the ampulla than the isthmus. In the ampulla, transcription of these genes was greater for the SF-SCL group, while the opposite was observed in the isthmus. The expression of the 88 most abundant miRNAs and 14 miRNAs in the ampulla and 34 miRNAs in isthmus were differentially expressed between LF-LCL and SF-SCL groups. Integration of transcriptomic and miRNA data and molecular pathways enrichment showed that important pathways were inhibited in the SF-SCL group due to miRNA control. In conclusion, the endocrine milieu affects the miRNA expression in the bovine oviduct in a region-specific manner.


Asunto(s)
Bovinos , Trompas Uterinas/efectos de los fármacos , Hormonas Esteroides Gonadales/farmacología , MicroARNs , Animales , Bovinos/genética , Bovinos/metabolismo , Ciclo Estral/efectos de los fármacos , Ciclo Estral/genética , Trompas Uterinas/metabolismo , Femenino , Perfilación de la Expresión Génica/veterinaria , Regulación de la Expresión Génica/efectos de los fármacos , MicroARNs/efectos de los fármacos , MicroARNs/genética , MicroARNs/metabolismo , Folículo Ovárico/efectos de los fármacos , Folículo Ovárico/metabolismo , Ovulación/efectos de los fármacos , Ovulación/genética , Procesamiento Postranscripcional del ARN/efectos de los fármacos , Procesamiento Postranscripcional del ARN/genética , Transcriptoma/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA