Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.590
Filtrar
1.
Reprod Biol ; 24(1): 100848, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38199161

RESUMO

Growth hormone is a key endocrine factor for metabolic adaptations to lactation and optimal reproductive function of the dairy cow. This study aimed to analyze the expression of GH and its receptor (GHR) in ovarian follicles, along with metabolic biomarkers, during the resumption of the postpartum follicular development, and to analyze the immunolocalization and protein expression of GH and GHR in preovulatory follicles. Thirty-six dairy cows were grouped according to the postpartum days (PPD) until the establishment of the first dominant follicle in: cows that established their first dominant follicle at fewer postpartum days (FPPD group; n = 15) and cows that established their first dominant follicle at more postpartum days (MPPD group; n = 22). For a second analysis, the same cows were regrouped according to the calving season (S), into cows calving in autumn (n = 20) and cows calving in winter (n = 17). During the PP, blood and follicular aspirates were obtained at two timepoints (T): when the first dominant follicle was established (T1, day 9 ± 2), and when the preovulatory follicle was established (T2, day 45 ± 2). Also, six dairy cows were ovariectomized in proestrus and ovarian histological sections were obtained. Growth hormone mRNA was detected in granulose cells from ovarian follicle sampled during PP. A PPD × T interaction was observed for GHR mRNA, where it was greater in the FPPD cows than in the MPPD cows at T1. Metabolic biomarkers and reproductive hormones showed differences or interaction between PPD, T, S, depending on the case. Also, GH and GHR were immunolocalized in granulosa and theca interna cells of preovulatory follicles. These results confirm the expression of GH and GHR in the mature ovarian follicles of dairy cows and show a possible association between greater GHR expression and an earlier resumption of postpartum follicular development.


Assuntos
Hormônio do Crescimento , Período Pós-Parto , Feminino , Humanos , Bovinos , Animais , Período Pós-Parto/fisiologia , Folículo Ovariano/fisiologia , Lactação/fisiologia , RNA Mensageiro , Biomarcadores , Ovulação/fisiologia
2.
Physiol Behav ; 275: 114451, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38176291

RESUMO

Early exposure of does to sexually active bucks triggers early puberty onset correlating with neuroendocrine changes. However, the sensory pathways that are stimulated by the male are still unknown. Here, we assessed whether responses to olfactory stimuli are modulated by social experience (exposure to males or not) and/or endocrine status (prepubescent or pubescent). We used a calcium imaging approach on goat sensory cells from the main olfactory epithelium (MOE) and the vomeronasal organ (VNO). For both cell types, we observed robust responses to active male hair in females under three physiological conditions: prepubescent females isolated from males (ISOL PrePub), pubescent females exposed to males (INT Pub) and isolated females (ISOL Pub). Response analysis showed overall greater proportion of responses to buck hair in ISOL PrePub. We hypothesized that females would be more responsive to active buck hair during the prepubertal period, with numerous responses perhaps originating from immature neurons. We also observed a greater proportion of mature olfactory neurons in the MOE and VNO of INT Pub females suggesting that male exposure can induce plastic changes on olfactory cell function and organization. To determine whether stimulation by male odor can advance puberty, we exposed prepubescent does to active buck hair (ODOR). In both ODOR and females isolated from males (ISOL) groups, puberty was reached one month after females exposed to intact bucks (INT), suggesting that olfactory stimulation is not sufficient to trigger puberty.


Assuntos
Ovulação , Comportamento Sexual Animal , Animais , Feminino , Masculino , Comportamento Sexual Animal/fisiologia , Estações do Ano , Ovulação/fisiologia , Olfato , Cabras/fisiologia
3.
Genetics ; 226(1)2024 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-38000906

RESUMO

Gamete development is a fundamental process that is highly conserved from early eukaryotes to mammals. As germ cells develop, they must coordinate a dynamic series of cellular processes that support growth, cell specification, patterning, the loading of maternal factors (RNAs, proteins, and nutrients), differentiation of structures to enable fertilization and ensure embryonic survival, and other processes that make a functional oocyte. To achieve these goals, germ cells integrate a complex milieu of environmental and developmental signals to produce fertilizable eggs. Over the past 50 years, Drosophila oogenesis has risen to the forefront as a system to interrogate the sophisticated mechanisms that drive oocyte development. Studies in Drosophila have defined mechanisms in germ cells that control meiosis, protect genome integrity, facilitate mRNA trafficking, and support the maternal loading of nutrients. Work in this system has provided key insights into the mechanisms that establish egg chamber polarity and patterning as well as the mechanisms that drive ovulation and egg activation. Using the power of Drosophila genetics, the field has begun to define the molecular mechanisms that coordinate environmental stresses and nutrient availability with oocyte development. Importantly, the majority of these reproductive mechanisms are highly conserved throughout evolution, and many play critical roles in the development of somatic tissues as well. In this chapter, we summarize the recent progress in several key areas that impact egg chamber development and ovulation. First, we discuss the mechanisms that drive nutrient storage and trafficking during oocyte maturation and vitellogenesis. Second, we examine the processes that regulate follicle cell patterning and how that patterning impacts the construction of the egg shell and the establishment of embryonic polarity. Finally, we examine regulatory factors that control ovulation, egg activation, and successful fertilization.


Assuntos
Oócitos , Oogênese , Animais , Feminino , Oogênese/genética , Oócitos/fisiologia , Ovulação/fisiologia , Folículo Ovariano , Drosophila , Mamíferos
4.
Psychoneuroendocrinology ; 160: 106669, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37988874

RESUMO

The cortisol awakening response (CAR) is influenced by several state and trait variables, one of which might be the menstrual cycle in women. Previous results suggested that the CAR is enhanced around ovulation, which is why it has been recommended to avoid sampling during the ovulatory phase. In two separate studies, we aimed to replicate previous findings that reported the CAR's modulation across the menstrual cycle, especially during ovulation. In Study 1, a group of 27 healthy naturally cycling women collected saliva at 0, 30, 45, and 60 min post-awakening on two days during their follicular, ovulatory, and luteal phases in a repeated measures design. In Study 2, CAR samples were collected from 30 healthy naturally cycling women on seven consecutive days around the expected ovulation. To increase reliability of CAR measurements, participants' compliance of saliva sampling times was monitored, ovarian steroids (estradiol and progesterone) were collected, and ovulation was confirmed with specific test kits. Contrary to our expectations, we detected no differences in the CAR over the menstrual cycle, and no significant association with variations in estradiol and progesterone. In addition, we excluded confounding effects such as compliance and validated the cycle phase. These results suggest that the CAR is largely robust against hormonal variations across the menstrual cycle, including the mid-cycle phase around ovulation. However, further research is needed to understand the potential ovarian steroid-induced modulation of HPA axis functioning and the menstrual cycle's effects on salivary cortisol levels in psychobiological studies.


Assuntos
Hidrocortisona , Progesterona , Feminino , Humanos , Progesterona/farmacologia , Hidrocortisona/farmacologia , Sistema Hipotálamo-Hipofisário/fisiologia , Reprodutibilidade dos Testes , Sistema Hipófise-Suprarrenal/fisiologia , Ovulação/fisiologia , Ciclo Menstrual/fisiologia , Estradiol/farmacologia , Esteroides/farmacologia , Saliva
5.
Biol Reprod ; 110(2): 288-299, 2024 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-37847612

RESUMO

Luteinizing hormone (LH) induces ovulation by acting on its receptors in the mural granulosa cells that surround a mammalian oocyte in an ovarian follicle. However, much remains unknown about how activation of the LH receptor modifies the structure of the follicle such that the oocyte is released and the follicle remnants are transformed into the corpus luteum. The present study shows that the preovulatory surge of LH stimulates LH receptor-expressing granulosa cells, initially located almost entirely in the outer layers of the mural granulosa, to rapidly extend inwards, intercalating between other cells. The cellular ingression begins within 30 min of the peak of the LH surge, and the proportion of LH receptor-expressing cell bodies in the inner half of the mural granulosa layer increases until the time of ovulation, which occurs at about 10 h after the LH peak. During this time, many of the initially flask-shaped cells appear to detach from the basal lamina, acquiring a rounder shape with multiple filipodia. Starting at about 4 h after the LH peak, the mural granulosa layer at the apical surface of the follicle where ovulation will occur begins to thin, and the basolateral surface develops invaginations and constrictions. Our findings raise the question of whether LH stimulation of granulosa cell ingression may contribute to these changes in the follicular structure that enable ovulation.


Assuntos
Hormônio Luteinizante , Receptores do LH , Feminino , Camundongos , Animais , Hormônio Luteinizante/metabolismo , Receptores do LH/metabolismo , Células da Granulosa/metabolismo , Folículo Ovariano/metabolismo , Ovulação/fisiologia , Mamíferos/metabolismo
6.
Biol Reprod ; 110(1): 130-139, 2024 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-37801701

RESUMO

The endocrinology regulating ovulation of the desired number of oocytes in the ovarian cycle is well described, particularly in mono-ovulatory species. Less is known about the characteristics that make one follicle suitable for ovulation while most other follicles die by atresia. Bromodeoxyuridine (BrdU) injection was used to characterize granulosa cell proliferation rates in developing ovarian follicles in the estrous cycle of mice. This methodology allowed identification of follicle diameters of secondary (80-130 µm), follicle-stimulating hormone (FSH)-sensitive (130-170 µm), FSH-dependent (170-350 µm), and preovulatory (>350 µm) follicles. Few preovulatory-sized follicles were present in the ovaries of mice at estrus, the beginning of the cycle. Progressive increases were seen at metestrus and diestrus, when full accumulation of the preovulatory cohort (~10 follicles) occurred. BrdU pulse-chase studies determined granulosa cell proliferation rates in the 24-48 h before the follicle reached the preovulatory stage. This showed that slow-growing follicles were not able to survive to the preovulatory stage. Mathematical modeling of follicle growth rates determined that the largest follicles at the beginning of the cycle had the greatest chance of becoming preovulatory. However, smaller follicles could enter the preovulatory follicle pool if low numbers of large antral follicles were present at the beginning of the cycle. In this instance, rapidly growing follicles had a clear selection advantage. The developing follicle pool displays heterogeneity in granulosa cell proliferation rates, even among follicles at the same stage of development. This parameter appears to influence whether a follicle can ovulate or become atretic.


Assuntos
Folículo Ovariano , Ovulação , Humanos , Feminino , Camundongos , Animais , Bromodesoxiuridina/metabolismo , Folículo Ovariano/metabolismo , Ovulação/fisiologia , Ovário , Hormônio Foliculoestimulante/farmacologia , Hormônio Foliculoestimulante/metabolismo
7.
Mol Cell Endocrinol ; 579: 112091, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-37863469

RESUMO

Female reproductive efficiency is influenced by the outcomes of various processes, including folliculogenesis, apoptosis, response to gonadotropin signaling, oocyte maturation, and ovulation. The role of hormones in regulating these processes and other reproductive activities has been well established. It is becoming increasingly evident that in addition to well-characterized hormones, growth factors play vital roles in regulating some of these reproductive activities. Growth factors and their receptors are widely distributed in vertebrate ovaries at different stages of ovarian development, indicating their involvement in intraovarian reproductive functions. In the ovary, cell surface receptors allow growth factors to regulate intraovarian reproductive activities. Understanding these actions in the reproductive axis would provide a tool to target growth factors and/or their receptors to yield desirable reproductive outcomes. These include enrichment of in vitro maturation and fertilization culture media, and management of infertility. This review discusses some widely characterized growth factors belonging to the TGF, EGF, IGF, FGF, and BDNF family of peptides and their role in female reproduction in vertebrates, with a focus on mammals.


Assuntos
Ovário , Ovulação , Animais , Feminino , Ovário/metabolismo , Ovulação/fisiologia , Gonadotropinas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Reprodução , Vertebrados , Oócitos/fisiologia , Mamíferos
8.
Sensors (Basel) ; 23(24)2023 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-38139575

RESUMO

(1) Background: Hormonal fluctuations across the menstrual cycle lead to multiple changes in physiological parameters such as body temperature, cardiovascular function, respiratory rate and perfusion. Electronic wearables analyzing those parameters might present a convenient alternative to urinary ovulation tests for predicting the fertile window. (2) Methods: We conducted a prospective observational study including women aged 18-45 years without current hormonal therapy who used a wrist-worn medical device and urinary ovulation tests for a minimum of three cycles. We analyzed the accuracy of both the retrospective and prospective algorithms using a generalized linear mixed-effects model. The findings were compared to real-world data from bracelet users who also reported urinary ovulation tests. (3) Results: A total of 61 study participants contributing 205 cycles and 6081 real-life cycles from 3268 bracelet users were included in the analysis. The mean error in identifying ovulation with the wrist-worn medical device retrospective algorithm in the clinical study was 0.31 days (95% CI -0.13 to 0.75). The retrospective algorithm identified 75.4% of fertile days, and the prospective algorithm identified 73.8% of fertile days correctly within the pre-specified equivalence limits (±2 days). The quality of the retrospective algorithm in the clinical study could be confirmed by real-world data. (4) Conclusion: Our data indicate that wearable sensors may be used to accurately detect the periovulatory period.


Assuntos
Ovulação , Punho , Feminino , Humanos , Estudos Retrospectivos , Estudos Prospectivos , Ovulação/fisiologia , Ciclo Menstrual/fisiologia
9.
Front Endocrinol (Lausanne) ; 14: 1282658, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38027176

RESUMO

Macrophages (MΦs) are the most abundant leukocytes in mammalian ovaries that have heterogeneity and plasticity. A body of evidence has indicated that these cells are important in maintaining ovarian homeostasis and they play critical roles in ovarian physiological events, such as folliculogenesis, ovulation, corpus luteum formation and regression. As females age, ovarian tissue microenvironment is typified by chronic inflammation with exacerbated ovarian fibrosis. In response to specific danger signals within aged ovaries, macrophages polarize into different M1 or M2 phenotypes, and specialize in unique functions to participate in the ovarian aging process. In this review, we will focus on the physiologic roles of MΦs in normal ovarian functions. Furthermore, we will discuss the roles of MΦs in the process of ovarian senescence, as well as the novel techniques applied in this field.


Assuntos
Ovário , Ovulação , Feminino , Animais , Ovário/fisiologia , Ovulação/fisiologia , Macrófagos , Leucócitos , Mamíferos
10.
Biol Reprod ; 109(6): 904-917, 2023 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-37712895

RESUMO

Cathepsin L plays physiological and pathological roles in immune responses, cancer, metamorphosis, and oogenesis in several species. However, the function of Cathepsin L in medaka ovaries remains unclear. Therefore, here, we examined the physiological functions of Cathepsin L in the medaka ovaries. Cathepsin L mRNA transcripts and proteins were found to be constitutively expressed in the ovaries of Oryzias latipes over a 24-h spawning cycle. Expression was localized within the oocyte cytoplasm of growing follicles and the follicle layer of preovulatory and postovulatory follicles. Moreover, the active form of Cathepsin L was highly expressed in the follicle layer of periovulatory follicles and the ovaries 2-6 h after ovulation. Recombinant Cathepsin L was activated under acidic conditions and exhibited enzymatic activity in acidic and neutral pH conditions. However, extracellular matrix proteins were degraded by recombinant Cathepsin L under acidic, not neutral pH conditions. Cathepsin L was secreted from preovulatory follicles, while active recombinant Cathepsin L was detected in the conditioned medium of a medaka cell line, OLHNI-2. Mechanistically, recombinant Cathepsin L activates recombinant urokinase-type plasminogen activator-1, which is expressed within the follicle layers post-ovulation. Meanwhile, the treatment of medakas with an E-64 or anti-Cathepsin L antibody effectively blocked follicular layer degeneration and degradation after ovulation, whereas in vitro ovulation was not inhibited by either. Collectively, the findings of this study indicate that although Cathepsin L does not impact ovulation in medakas, it contributes to the degeneration and degradation of the follicle layers following ovulation via activation of urokinase-type plasminogen activator-1, and not via the degradation of extracellular matrix proteins.


Assuntos
Oryzias , Ovário , Feminino , Animais , Ovário/fisiologia , Oryzias/fisiologia , Catepsina L/genética , Catepsina L/farmacologia , Ativador de Plasminogênio Tipo Uroquinase/farmacologia , Ovulação/fisiologia , Proteínas da Matriz Extracelular
11.
Acta Obstet Gynecol Scand ; 102(7): 914-920, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37194424

RESUMO

INTRODUCTION: We aimed to describe physiological changes in endometrial blood flow (minute arterioles running through the endometrium) from ovulation to the mid-luteal phase using superb microvascular imaging. MATERIAL AND METHODS: The study involved 17 women (median age, 32.5 years; first to third interquartile range, 29.8-40.0 years) with regular menstrual cycles who were managed in our institute from 2020 to 2021. The uterus was delineated at the sagittal section using transvaginal ultrasonography incorporated with superb microvascular imaging. For each participant, a total of 28 cycles were observed; 17 cycles observed within one day of ovulation and the implantation period, 5-7 days (D5-7) after ovulation in the same cycle, and nine cycles in which only ovulation was observed, and two cycles in which only D5-7 was observed. Therefore, 26 and 19 images at ovulation and D5-7, respectively, were acquired. Endometrial blood flow was evaluated by depth of the vascular signal in the endometrium and categorized as follows: signals only in the basal layer of the endometrium (grade 1), reaching up to half the endometrium (grade 2), and covering the whole endometrium (grade 3). Changes in the grade of endometrial blood flow from ovulation to D5-7 after ovulation, and the relationship between the grade of endometrial blood flow and the endometrial thickness on ovulation and D5-7 after ovulation, were analyzed. Statistical significance was set at p < 0.05. RESULTS: The endometrial blood flow from ovulation to D5-7 after ovulation during the same menstrual period showed a downgrade in 14 of 17 cycles (82.3%) and no change in the remaining three cycles (17.6%), indicating a decrease in the endometrial blood flow from ovulation to D5-7 after ovulation (p = 0.001). There were differences between the grade of endometrial blood flow and median endometrial thickness on ovulation (grade 1: 5.9 mm, grade 2: 9.1 mm, and grade 3: 11.2 mm); however, no differences in the endometrial thickness were found between the grades on D5-7 after ovulation. CONCLUSIONS: In the normal menstrual cycle, endometrial blood flow decreased from ovulation to the mid-luteal phase, and the endometrial thickness in the ovulatory phase was related to the endometrial perfusion.


Assuntos
Endométrio , Fase Luteal , Feminino , Humanos , Adulto , Endométrio/diagnóstico por imagem , Ovulação/fisiologia , Ciclo Menstrual/fisiologia , Útero/irrigação sanguínea
12.
Gen Comp Endocrinol ; 338: 114280, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37011766

RESUMO

For endangered species managed ex situ, production of offspring is a key factor to ensure healthy and self-sustaining populations. However, current breeding goals for the whooping crane (Grus americana) are impeded by poor reproduction. Our study sought to better understand mechanisms regulating ovarian function in ex situ managed whooping cranes and the regulatory function of the hypothalamic-pituitary-gonadal (HPG) axis in relation to follicle formation and egg laying. To characterize hormonal regulation of follicular development and ovulation, we collected weekly blood samples from six female whooping cranes during two breeding seasons, for a total of 11 reproductive cycles. The plasma samples were assessed for follicle stimulating hormone, luteinizing hormone, estradiol, and progesterone and the yolk precursors vitellogenin and very low-density lipoprotein. Ultrasonographic examination of the ovary was conducted at the time of blood collection. Preovulatory follicles (>12 mm) were present in laying cycles (n = 6) but absent in non-laying cycles (n = 5). The patterns of plasma hormone and yolk precursor concentrations corresponded to the stage of follicle development. Specifically, gonadotropin and yolk precursor concentrations increased as follicles transitioned from the non-yolky to yolky stage but did not increase further as the follicle advanced to preovulatory and ovulatory stages. Estrogen and progesterone concentrations increased as follicle size increased and reached peak concentrations (P < 0.05) when follicles developed to ovulatory and preovulatory stages, respectively. While overall mean circulating gonadotropin, progesterone, and yolk precursor concentrations did not differ for laying versus non-laying cycles, mean plasma estradiol in laying cycles was significantly higher than that in non-laying cycles. In summary, the findings suggested that disruption of mechanisms regulating follicle recruitment is likely responsible for the oviposition failure of the captive female whooping crane.


Assuntos
Ovário , Progesterona , Animais , Feminino , Ovário/fisiologia , Aves , Hormônio Luteinizante , Estradiol , Hormônio Foliculoestimulante , Ovulação/fisiologia
13.
FASEB J ; 37(4): e22858, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36943419

RESUMO

The role of prostaglandins (PGs) in the ovulatory process is known. However, the role of the ATP binding cassette subfamily C member 4 (ABCC4), transmembrane PG carrier protein, in ovulation remains unknown. We report herein that ABCC4 expression is significantly upregulated in preovulatory human granulosa cells (GCs). We found that PGE2 efflux in cultured human GCs is mediated by ABCC4 thus regulating its extracellular concentration. The ABCC4 inhibitor probenecid demonstrated effective blocking of ovulation and affects key ovulatory genes in female mice in vivo. We postulate that the reduction in PGE2 efflux caused by the inhibition of ABCC4 activity in GCs decreases the extracellular concentration of PGE2 and its ovulatory effect. Treatment of female mice with low dose of probenecid as well as with the PTGS inhibitor indomethacin or Meloxicam synergistically blocks ovulation. These results support the hypothesis that ABCC4 has an important role in ovulation and might be a potential target for non-hormonal contraception, especially in combination with PGE2 synthesis inhibitors. These findings may fill the gap in understanding the role of ABCC4 in PGE2 signaling, enhance the understanding of ovulatory disorders, and facilitate the treatment and control of fertility.


Assuntos
Anticoncepcionais , Dinoprostona , Humanos , Feminino , Camundongos , Animais , Dinoprostona/metabolismo , Anticoncepcionais/metabolismo , Anticoncepcionais/farmacologia , Probenecid/metabolismo , Probenecid/farmacologia , Folículo Ovariano/metabolismo , Ovulação/fisiologia , Proteínas de Membrana Transportadoras/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo
14.
Biol Reprod ; 108(4): 629-644, 2023 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-36708230

RESUMO

Ovulation is an integral part of women's menstrual cycle and fertility. Understanding the mechanisms of ovulation has broad implications for the treatment of anovulatory diseases and the development of novel contraceptives. Now, few studies have developed effective models that both faithfully recapitulate the hallmarks of ovulation and possess scalability. We established a three-dimensional encapsulated in vitro follicle growth (eIVFG) system that recapitulates folliculogenesis and produces follicles that undergo ovulation in a controlled manner. Here, we determined whether ex vivo ovulation preserves molecular signatures of ovulation and demonstrated its use in discovering novel ovulatory pathways and nonhormonal contraceptive candidates through a high-throughput ovulation screening. Mature murine follicles from eIVFG were induced to ovulate ex vivo using human chorionic gonadotropin and collected at 0, 1, 4, and 8 hours post-induction. Phenotypic analyses confirmed key ovulatory events, including cumulus expansion, oocyte maturation, follicle rupture, and luteinization. Single-follicle RNA-sequencing analysis revealed the preservation of ovulatory genes and dynamic transcriptomic profiles and signaling. Soft clustering identified distinct gene expression patterns and new pathways that may critically regulate ovulation. We further used this ex vivo ovulation system to screen 21 compounds targeting established and newly identified ovulatory pathways. We discovered that proprotein convertases activate gelatinases to sustain follicle rupture and do not regulate luteinization and progesterone secretion. Together, our ex vivo ovulation system preserves molecular signatures of ovulation, presenting a new powerful tool for studying ovulation and anovulatory diseases as well as for establishing a high-throughput ovulation screening to identify novel nonhormonal contraceptives for women.


Assuntos
Anovulação , Anticoncepcionais , Feminino , Humanos , Animais , Camundongos , Anticoncepcionais/farmacologia , Ovulação/fisiologia , Folículo Ovariano/metabolismo , Oogênese , Ciclo Menstrual , Progesterona/farmacologia
15.
Reprod Domest Anim ; 58(1): 48-59, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36102495

RESUMO

The objectives of the experiment were to determine the effect of two doses of equine chorionic gonadotropin (eCG) in a standard synchronization protocol based on a short-term progesterone (P4 ) priming on ovarian structures and haemodynamics, concentrations of steroid hormones and prolificacy rate when oestrus was induced during low-breeding season (LBS) in Beetal dairy goats. We hypothesized that inclusion of eCG in a short-term P4 priming-based synchronization protocol would increase the blood perfusion to ovarian structures leading to enhance oestrous and ovulatory responses and prolificacy rate in goats. Forty-two multiparous acyclic goats were blocked by body condition and, within block, assigned randomly to receive saline as control (CON), low eCG (L-eCG; 300 IU) or high eCG (H-eCG; 600 IU) dose. Initially, a controlled internal drug release (CIDR) device was placed in the anterior vagina on d -8, followed by removal of CIDR on d -3, concurrent with the administration of PGF2α and eCG according to their respective treatments. Goats were monitored for oestrous response. B-mode and Doppler ultrasonography was performed with 12-h interval, starting from day -3 until natural breeding (day 0), and then on days 5, 10, 15 and 20 post-breeding to monitor follicular and luteal dynamics and blood flow, respectively. Blood was sampled at 0, 12, 24, 36 and 60 h after CIDR removal to quantify plasma concentrations of estradiol-17ß (E2 ), whereas plasma concentrations of P4 were assayed at days 5, 10, 15 and 20 after breeding. Pregnancy and prolificacy rates were determined at day 30 and 150 after breeding, respectively. Data were analysed with mixed-effects models, and orthogonal contrasts were used to evaluate the effect of treatment [Con vs. (½ L-eCG + ½ H-eCG)] and dose of eCG (L-eCG vs. H-eCG). Data are presented in sequence as CON, L-eCG, H-eCG (LSM ± SEM). The oestrous intensity score (152.9 vs. 182.7 vs. 186.5 ± 15.1; p = .02) was greater in eCG-treated goats as compared to CON. Administration of eCG reduced the intervals to standing oestrus (66.2 vs. 41.8 vs. 48.9 h ± 5.5; p = .05), breeding (70.2 vs. 44.4 vs. 45.4 h ± 4.5; p = .03) and ovulation (84.5 vs. 61.2 vs. 63.4 h ± 6.2; p = .05) compared with CON goats. The mean growth rate of pre-ovulatory follicle was greater (1.11 vs. 1.49 vs. 1.45 mm ± 0.08; p = .01) in eCG-treated goats resulting in an increased diameter of pre-ovulatory follicle (6.27 vs. 7.20 vs. 7.31 mm ± 0.07; p < .01) and corpora lutea (6.75 vs. 8.26 vs. 8.07 mm ± 0.42; p = .04) than CON. The mean follicular blood flow did not differ among treatments; however, the mean luteal blood flow was greater in L-eCG-treated goats (0.81 vs. 1.61 vs. 1.07 cm2 ± 0.12; p = .001). The mean concentrations of E2 (4.03 vs. 5.21 vs. 4.78 pg/ml ± 0.42; p = .04) and P4 (4.85 vs. 6.39 vs. 6.22 ng/ml ± 0.34; p = .04) were greater in eCG-treated goats. The twinning rate did not differ between treatments; nevertheless, prolificacy rate was greater (p = .04) in L-eCG-treated goats. Collectively, our data suggest that the administration of eCG improves the induction of oestrous and ovarian dynamics. Administration of L-eCG enhances prolificacy rate, therefore, a low dose of eCG might be practically beneficial to improve reproduction during LBS in acyclic Beetal dairy goats.


Assuntos
Sincronização do Estro , Cabras , Gravidez , Feminino , Animais , Cavalos , Estações do Ano , Cabras/fisiologia , Sincronização do Estro/métodos , Progesterona , Ovulação/fisiologia , Estradiol , Hemodinâmica
16.
Biol Reprod ; 108(2): 258-268, 2023 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-36214501

RESUMO

Angiogenesis within the ovarian follicle is an important component of ovulation. New capillary growth is initiated by the ovulatory surge of luteinizing hormone (LH), and angiogenesis is well underway at the time of follicle rupture. LH-stimulated follicular production of vascular growth factors has been shown to promote new capillary formation in the ovulatory follicle. The possibility that LH acts directly on ovarian endothelial cells to promote ovulatory angiogenesis has not been addressed. For these studies, ovaries containing ovulatory follicles were obtained from cynomolgus macaques and used for histological examination of ovarian vascular endothelial cells, and monkey ovarian microvascular endothelial cells (mOMECs) were enriched from ovulatory follicles for in vitro studies. mOMECs expressed LHCGR mRNA and protein, and immunostaining confirmed LHCGR protein in endothelial cells of ovulatory follicles in vivo. Human chorionic gonadotropin (hCG), a ligand for LHCGR, increased mOMEC proliferation, migration and capillary-like sprout formation in vitro. Treatment of mOMECs with hCG increased cAMP, a common intracellular signal generated by LHCGR activation. The cAMP analog dibutyryl cAMP increased mOMEC proliferation in the absence of hCG. Both the protein kinase A (PKA) inhibitor H89 and the phospholipase C (PLC) inhibitor U73122 blocked hCG-stimulated mOMEC proliferation, suggesting that multiple G-proteins may mediate LHCGR action. Human ovarian microvascular endothelial cells (hOMECs) enriched from ovarian aspirates obtained from healthy oocyte donors also expressed LHCGR. hOMECs also migrated and proliferated in response to hCG. Overall, these findings indicate that the LH surge may directly activate ovarian endothelial cells to stimulate angiogenesis of the ovulatory follicle.


Assuntos
Células Endoteliais , Neovascularização Fisiológica , Ovário , Receptores do LH , Animais , Feminino , Humanos , Gonadotropina Coriônica/farmacologia , Gonadotropina Coriônica/metabolismo , Células Endoteliais/metabolismo , Hormônio Luteinizante/farmacologia , Hormônio Luteinizante/metabolismo , Macaca fascicularis , Neovascularização Fisiológica/fisiologia , Folículo Ovariano/metabolismo , Ovário/irrigação sanguínea , Ovário/metabolismo , Ovulação/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Receptores do LH/genética , Receptores do LH/metabolismo
17.
Mol Cell Endocrinol ; 560: 111816, 2023 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-36410550

RESUMO

We have previously shown that the prostaglandin E2/Ptger4b receptor system is involved in ovulation in teleost medaka and induces intracellular actin cytoskeleton rearrangement in the granulosa cells of preovulatory follicles. In this study, we investigated the signaling pathways through which prostaglandin E2 induces a change in the actin cytoskeleton. Treating preovulatory follicles with GW627368X (Ptger4b antagonist), a Rho inhibitor, or Y-27632 [Rho-associated protein kinase (Rock) inhibitor] inhibited not only in vitro follicle ovulation but also intracellular actin cytoskeleton rearrangement. Active Rhoa-c and Rock1 were detected in follicles immediately before ovulation. GW627368X also inhibited Rhoa-c activation and cytoskeleton rearrangement. PGE2-induced actin cytoskeleton rearrangement was not observed in the Ptger4b-, Rhoa-c-, or Rock1-deficient OLHNI-2 cells. These results indicate that the PGE2/Ptger4b pathway regulates intracellular actin cytoskeleton rearrangement via the Rho/Rock pathway in the granulosa cells of preovulatory follicles during medaka ovulation.


Assuntos
Oryzias , Feminino , Animais , Dinoprostona , Ovulação/fisiologia , Células da Granulosa , Citoesqueleto de Actina , Quinases Associadas a rho
18.
Theriogenology ; 197: 31-36, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36463668

RESUMO

Progesterone treatment for synchrony of estrus is standard in sheep artificial insemination (AI) programs but can be associated with poor outcomes. Potential for improvement exists through a better understanding of the interactions between follicle development, luteal regression, emergence of the ovulatory follicle and timing of estrus. These interactions were examined by comparing progesterone-treated (Day 1 = day of pessary insertion) and naturally cycling ewes (Day 1 = day after estrus) at three times of the year (Autumn, Spring equinox and late Spring). Observations were made from Day 1 until the day of ovulation. Compared with the natural cycle, progesterone treatment (300 mg intra-vaginal pessary for 14 d) reduced the number of follicle waves (2.2 ± 0.18 versus 2.8 ± 0.12; P < 0.05) and increased the length of the ovulatory wave (8.6 ± 0.45 versus 6.6 ± 0.42 d; P < 0.05). The number of follicles per wave, the inter-wave interval and ovulation rate were not affected. However, progesterone treatment induced (P < 0.05) an earlier luteolysis (9.7 ± 0.51 versus 15.4 ± 0.49 d after Day 1), an earlier emergence of the ovulatory follicle (7.5 ± 0.48 versus 11.4 ± 0.46 d after Day 1) and an earlier onset of estrus (26.1 ± 2.95 versus 53.3 ± 2.84 h after Day 14). Time of year also influenced the response to progesterone treatment. In Autumn compared with the Spring equinox and late Spring, there was a reduction (P < 0.05) in follicle wave number (2.4 ± 0.21 versus 2.5 ± 0.29 versus 3.0 ± 0.20 respectively), follicles per wave (2.6 ± 0.27 versus 3.5 ± 0.25 versus 3.2 ± 0.20 respectively), ovulation rate (1.6 ± 0.12 versus 1.9 ± 0.12 versus 2.0 ± 0.10 respectively) and the inter-wave interval was longer (5.3 ± 0.40 versus 4.0 ± 0.32 versus 3.8 ± 0.27 d respectively; P < 0.05). Time of year also influenced (P < 0.05) the time of luteolysis (earliest in late Spring), emergence of the ovulatory follicle (earliest in Autumn) and onset of estrus (earliest in Autumn). It is concluded that (1) the effects of progesterone treatment on follicle waves are relatively minor, (2) the effects of treatment on timing of luteolysis, emergence of the ovulatory follicle and onset of estrus are all significant although the effects on AI outcomes remain to be determined and (3) time of year has a minimal effect on follicle waves but a more significant effect on other parameters of the estrous cycle. A better understanding of these complexities will assist in the development of improved protocols for synchrony of estrus.


Assuntos
Pessários , Progesterona , Feminino , Animais , Ovinos , Progesterona/farmacologia , Folículo Ovariano/fisiologia , Ovulação/fisiologia , Ciclo Estral , Estradiol/farmacologia , Ultrassonografia
19.
Biol Reprod ; 108(1): 107-120, 2023 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-36345168

RESUMO

The luteinizing hormone (LH) surge induces paracrine mediators within the ovarian follicle that promote ovulation. The present study explores neurotensin (NTS), a neuropeptide, as a potential ovulatory mediator in the mouse ovary. Ovaries and granulosa cells (GCs) were collected from immature 23-day-old pregnant mare serum gonadotropin primed mice before (0 h) and after administration of human chorionic gonadotropin (hCG; an LH analog) across the periovulatory period (4, 8, 12, and 24 h). In response to hCG, Nts expression rapidly increased 250-fold at 4 h, remained elevated until 8 h, and decreased until 24 h. Expression of Nts receptors for Ntsr1 remained unchanged across the periovulatory period, Ntsr2 was undetectable, whereas Sort1 expression (also called Ntsr3) gradually decreased in both the ovary and GCs after hCG administration. To better understand Nts regulation, inhibitors of the LH/CG signaling pathways were utilized. Our data revealed that hCG regulated Nts expression through the protein kinase A (PKA) and p38 mitogen-activated protein kinase (p38MAPK) signaling pathways. Additionally, epidermal-like-growth factor (EGF) receptor signaling also mediated Nts induction in GCs. To elucidate the role of NTS in the ovulatory process, we used a Nts silencing approach (si-Nts) followed by RNA-sequencing (RNA-seq). RNA-seq analysis of GCs collected after hCG with or without si-Nts identified and qPCR confirmed Ell2, Rsad2, Vps37a, and Smtnl2 as genes downstream of Nts. In summary, these findings demonstrate that hCG induces Nts and that Nts expression is mediated by PKA, p38MAPK, and EGF receptor signaling pathways. Additionally, NTS regulates several novel genes that could potentially impact the ovulatory process.


Assuntos
Neurotensina , Ovário , Ovulação , Animais , Feminino , Camundongos , Gonadotropina Coriônica/farmacologia , Gonadotropina Coriônica/metabolismo , Células da Granulosa/metabolismo , Cavalos , Hormônio Luteinizante/metabolismo , Neurotensina/genética , Neurotensina/metabolismo , Folículo Ovariano/metabolismo , Ovário/metabolismo , Ovulação/genética , Ovulação/fisiologia , Fatores de Elongação da Transcrição/metabolismo
20.
Reproduction ; 165(2): 147-157, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36342662

RESUMO

In brief: In the proestrus day, the neural and endocrine signals modulate ovarian function. This study shows vagus nerve plays a role in the multisynaptic pathways of communication between the suprachiasmatic nucleus and the ovaries where such neural information determines ovulation. Abstract: The suprachiasmatic nucleus (SCN) regulates the activity of several peripheral organs through a parasympathetic-sympathetic pathway. Previously, we demonstrated that atropine (ATR) microinjection in the right SCN of rats during proestrus blocks ovulation. In the present study, we analysed whether the vagus nerve is one of the neural pathways by which the SCN regulates ovulation. For this, CIIZ-V strain cyclic rats on the day of proestrus were microinjected with a saline solution (vehicle) or ATR in the right or left SCN, which was followed by ventral laparotomy or ipsilateral vagotomy to the microinjection side. Some animal groups were sacrificed (i) on the same day of the surgery to measure oestradiol, progesterone and luteinizing hormone (LH) levels or (ii) at 24 h after surgery to evaluate ovulation. The left vagotomy in rats microinjected with ATR in the left SCN did not modify ovulation. In rats with ATR microinjection in the right SCN, the right vagotomy increased the levels of steroids and LH on the proestrus and ovulatory response. The present results suggest that the right vagus nerve plays a role in the multisynaptic pathways of communication between the SCN and the ovaries and indicate that such neural information participates in the regulation of the oestradiol and progesterone surge, which triggers the preovulatory peak of LH and determines ovulation.


Assuntos
Hormônio Luteinizante , Progesterona , Feminino , Ratos , Animais , Progesterona/metabolismo , Hormônio Luteinizante/metabolismo , Núcleo Supraquiasmático/metabolismo , Ovulação/fisiologia , Estradiol/metabolismo , Atropina/farmacologia , Atropina/metabolismo , Nervo Vago/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...