Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Reproduction ; 157(1): 13-25, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30394704

RESUMEN

A number of studies have demonstrated effects of gestational undernutrition on fetal ovarian development and postnatal female fertility. However, the mechanism underlying these effects remains elusive. Using a cohort of animals in which altered gestational nutrition affected indicators of postnatal fertility, this study applies RNAseq to fetal ovaries to identify affected genes and pathways that may underlie the relationship between gestational plane of nutrition and postnatal fertility. Pregnant ewes were exposed to either a maintenance diet or 0.6 of maintenance for the first 55 days of gestation followed by an ad libitum diet. Complementary DNA libraries were constructed from 5 to 6 fetal ovaries from each nutritional group at both days 55 and 75 of gestation and sequenced using Ion Proton. Of approximately 16,000 transcripts, 69 genes were differentially expressed at day 55 and 145 genes differentially expressed at day 75. At both gestational ages, genes expressed preferentially in germ cells were common among the differentially expressed genes. Enriched gene ontology terms included ion transport, nucleic acid binding, protease inhibitor activity and carrier proteins of the albumin family. Affected pathways identified by IPA analysis included LXR/RXR activation, FXR/RXR activation, pathways associated with nitric oxide production and citrullination (by NOS1), vitamin C transport and metabolism and REDOX reactions. The data offer some insights into potential mechanisms underlying the relationship between gestational plane of nutrition and postnatal fertility observed in these animals. In particular, the roles of nitric oxide and protease inhibitors in germ cell development are highlighted and warrant further study.


Asunto(s)
Feto/metabolismo , Desnutrición/genética , Ovario/embriología , Ovario/metabolismo , Fenómenos Fisiologicos de la Nutrición Prenatal , Ovinos , Animales , Femenino , Desarrollo Fetal/genética , Feto/embriología , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Desnutrición/metabolismo , Embarazo , Complicaciones del Embarazo/genética , Complicaciones del Embarazo/metabolismo , Fenómenos Fisiologicos de la Nutrición Prenatal/genética , Ovinos/embriología , Ovinos/genética , Ovinos/metabolismo
2.
J Morphol ; 279(11): 1603-1614, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30397936

RESUMEN

Light microscopy studies of the female American lobster Homarus americanus reproductive system are essentially nonexistent or outdated. Based on samples taken in the spring, summer, and autumn from the southern Gulf of St. Lawrence between 1994 and 2014, and using a combination of histological and scanning electron microscope techniques, we propose an ovarian cycle with 10 stages, identifying for the first time a recovery stage. Also, an atypical resorption stage, characterized by massive reabsorption of mature oocytes, is occasionally observed during summer months. The oviducts are composed of connective tissue (elastic and collagen fibers) with no muscle or secretory activities. Their epithelium shows a cyclic pattern and phagocytosis activities linked to spawning. Although the role of the seminal receptacle is to store and protect semen, free spermatozoa (i.e., without the spermatophoric wall and the acellular gelatinous substance that constitute the semen) were also observed in its posteriolateral grooves immediately prior to spawning, which is consistent with an external fertilization mechanism at the seminal receptacle. Unexpectedly, free spermatozoa were observed externally near two pore-like structures located on the gonopore's operculum, not at the seminal receptacle, after spawning; hence, more work is needed to fully understand the fertilization mechanism for the American lobster.


Asunto(s)
Genitales Femeninos/anatomía & histología , Genitales Femeninos/fisiología , Nephropidae/anatomía & histología , Nephropidae/fisiología , Animales , Femenino , Genitales Femeninos/ultraestructura , Nephropidae/ultraestructura , Oogénesis , Ovario/citología , Ovario/embriología
3.
Gen Comp Endocrinol ; 266: 60-66, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-29753927

RESUMEN

Spexin (Spx), a novel neuropeptide, composed of 14 amino acid residues, is evolutionally conserved from fish to mammals. It has been suggested that Spx has pleiotropic functions in mammals. However, reports about Spx are very limited. To clarify the roles of Spx in the regulation of reproduction and food-intake in the spotted scat, the spx (ssspx) gene was cloned and analyzed. Analysis of the tissue distribution by RT-PCR showed that ssspx expression was widespread. During ovary development, expression of ssspx was found to be highest in phase II, moderate in phase III, and at its lowest level in phase IV. Ssspx expression was significantly down-regulated in the hypothalamus after treatment with E2 both in vitro and in vivo. A significant increase of ssspx was observed after 2 and 7 days of food deprivation. However, the ssspx transcript levels in the 7 day fasting group decreased significantly after refeeding 3 h after the scheduled feeding time. This suggests that ssSpx may be involved in the regulation of reproduction and food-intake in the spotted scat.


Asunto(s)
Perfilación de la Expresión Génica , Hormonas Peptídicas/genética , Perciformes/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Clonación Molecular , Estradiol/farmacología , Ayuno , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Ovario/efectos de los fármacos , Ovario/embriología , Ovario/metabolismo , Hormonas Peptídicas/química , Hormonas Peptídicas/metabolismo , Perciformes/metabolismo , Filogenia , Reproducción , Alineación de Secuencia , Distribución Tisular/efectos de los fármacos
4.
PLoS One ; 12(5): e0177932, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28531178

RESUMEN

Uranium is an actinide naturally found in the environment. Anthropogenic activities lead to the release of increasing amounts of uranium and depleted uranium (DU) in the environment, posing potential risks to aquatic organisms due to radiological and chemical toxicity of this radionucleide. Although environmental contaminations with high levels of uranium have already been observed, chronic exposures of non-human species to levels close to the environmental quality standards remain scarcely characterized. The present study focused on the identification of the molecular pathways impacted by a chronic exposure of zebrafish to 20 µg/L of DU during 10 days. The transcriptomic effects were evaluated by the use of the mRNAseq analysis in three organs of adult zebrafish, the brain the testis and the ovaries, and two developmental stages of the adult fish progeny, two-cells embryo and four-days larvae. The results highlight generic effects on the cell adhesion process, but also specific transcriptomic responses depending on the organ or the developmental stage investigated. The analysis of the transgenerational effects of DU-exposure on the four-day zebrafish larvae demonstrate an induction of genes involved in oxidative response (cat, mpx, sod1 and sod2), a decrease of expression of the two hatching enzymes (he1a and he1b), the deregulation of the expression of gene coding for the ATPase complex and the induction of cellular stress. Electron microscopy analysis of skeletal muscles on the four-days larvae highlights significant histological impacts on the ultrastructure of both the mitochondria and the myofibres. In addition, the comparison with the transcriptomic data obtained for the acetylcholine esterase mutant reveals the induction of protein-chaperons in the skeletal muscles of the progeny of fish chronically exposed to DU, pointing towards long lasting effects of this chemical in the muscles. The results presented in this study support the hypothesis that a chronic parental exposure to an environmentally relevant concentration of DU could impair the progeny development with significant effects observed both at the molecular level and on the histological ultrastructure of organs. This study provides a comprehensive transcriptomic dataset useful for ecotoxicological studies on other fish species at the molecular level. It also provides a key DU responsive gene, egr1, which may be a candidate biomarker for monitoring aquatic pollution by heavy metals.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Análisis de Secuencia de ARN/métodos , Uranio/toxicidad , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Animales , Encéfalo/efectos de los fármacos , Encéfalo/embriología , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Masculino , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/embriología , Músculo Esquelético/ultraestructura , Ovario/efectos de los fármacos , Ovario/embriología , Testículo/efectos de los fármacos , Testículo/embriología , Contaminantes Radiactivos del Agua/toxicidad , Pez Cebra/embriología
5.
Endocrinology ; 157(4): 1535-45, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26741195

RESUMEN

Vitamin D (VitD) deficiency affects more than 1 billion people worldwide with a higher prevalence in reproductive-aged women and children. The physiological effects of maternal VitD deficiency on the reproductive health of the offspring has not been studied. To determine whether maternal VitD deficiency affects reproductive physiology in female offspring, we monitored the reproductive physiology of C57BL/6J female offspring exposed to diet-induced maternal VitD deficiency at three specific developmental stages: 1) in utero, 2) preweaning, or 3) in utero and preweaning. We hypothesized that exposure to maternal VitD deficiency disrupts reproductive function in exposed female offspring. To test this hypothesis, we assessed vaginal opening and cytology and ovary and pituitary function as well as gonadotropin and gonadal steroid levels in female offspring. The in utero, preweaning, and in utero and preweaning VitD deficiency did not affect puberty. However, all female mice exposed to maternal VitD deficiency developed prolonged and irregular estrous cycles characterized by oligoovulation and extended periods of diestrus. Despite similar gonadal steroid levels and GnRH neuron density, females exposed to maternal VitD deficiency released less LH on the evening of proestrus. When compared with control female offspring, there was no significant difference in the ability of females exposed to maternal VitD deficiency to respond robustly to exogenous GnRH peptide or controlled ovarian hyperstimulation. These findings suggest that maternal VitD deficiency programs reproductive dysfunction in adult female offspring through adverse effects on hypothalamic function.


Asunto(s)
Sistemas Neurosecretores/fisiopatología , Complicaciones del Embarazo/fisiopatología , Reproducción/fisiología , Deficiencia de Vitamina D/fisiopatología , Animales , Animales Recién Nacidos , Femenino , Hipotálamo/embriología , Hipotálamo/crecimiento & desarrollo , Hipotálamo/fisiopatología , Hormona Luteinizante/sangre , Hormona Luteinizante/metabolismo , Masculino , Ratones Endogámicos C57BL , Sistemas Neurosecretores/embriología , Sistemas Neurosecretores/crecimiento & desarrollo , Ovario/embriología , Ovario/crecimiento & desarrollo , Ovario/fisiopatología , Hipófisis/embriología , Hipófisis/crecimiento & desarrollo , Hipófisis/fisiopatología , Embarazo , Maduración Sexual/fisiología , Factores de Tiempo , Destete
6.
Gen Comp Endocrinol ; 221: 134-43, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25701739

RESUMEN

Estradiol-17ß (E2) and maturation-inducing hormone (MIH) are two steroid hormones produced in the teleost ovary that are required for vitellogenic growth and final oocyte maturation and ovulation. During this transition, the main steroid hormone produced in the ovary shifts from estrogens to progestogens. In the commercially important Japanese eel (Anguilla japonica), the MIH 17α,20ß-dihydroxy-4-pregnen-3-one (DHP) is generated from its precursor by P450c17, which has both 17α-hydroxylase and C17-20 lyase activities. In order to elucidate the regulatory mechanism underlying the steroidogenic shift from E2 to DHP and the mechanistic basis for the failure of this shift in artificially matured eels, the cDNA for cyp17a2-which encodes P450c17-II-was isolated from the ovary of wild, mature Japanese eel and characterized, and the expression patterns of cyp17a1 and cyp17a2 during induced ovarian development were investigated in cultured eel ovaries. Five cDNAs (types I-V) encoding P450c17-II were identified that had minor sequence variations. HEK293T cells transfected with all but type II P450c17-II converted exogenous progesterone to 17α-hydroxyprogesterone (17α-P), providing evidence for 17α-hydroxylase activity; however, a failure to convert 17α-P to androstenedione indicated that C17-20 lyase activity was absent. Cyp17a2 mRNA was expressed mainly in the head kidney, ovary, and testis, and quantitative PCR analysis demonstrated that expression in the ovary increased during induced vitellogenesis and oocyte maturation/ovulation. In contrast, P450c17-I showed both 17α-hydroxylase and C17-20 lyase activities, and cyp17a1 expression increased until the mid-vitellogenic stage and remained high thereafter. Considering the high level of cyp17a2 transcript in the eel ovary at the migratory nucleus stage together with our previous report demonstrating that eel ovaries have strong 17α-P-to-DHP conversion activity, the failure of artificially maturing eels to produce the maturation-inducing DHP may be explained by a deficiency in 17α-P production due to the persistence of cyp17a1 expression after the completion of vitellogenesis.


Asunto(s)
Anguilla/metabolismo , ADN Complementario/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Ovario/embriología , Esteroide 17-alfa-Hidroxilasa/genética , Vitelogénesis/fisiología , Secuencia de Aminoácidos , Anguilla/genética , Anguilla/crecimiento & desarrollo , Animales , Secuencia de Bases , Estradiol/metabolismo , Femenino , Células HEK293 , Humanos , Masculino , Datos de Secuencia Molecular , Progesterona/metabolismo , Progestinas/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico , Factores Sexuales , Esteroide 17-alfa-Hidroxilasa/metabolismo , Esteroides/metabolismo , Testículo/metabolismo
7.
Dev Biol ; 381(1): 144-58, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23769843

RESUMEN

Sex determination in mammals requires interaction between the transcription factor GATA4 and its cofactor FOG2. We have recently described the function of both proteins in testis development beyond the sex determination stage; their roles in the postnatal ovary, however, remain to be defined. Here, we use gene targeting in mice to determine the requirement of GATA4 and FOG2 in ovarian development and folliculogenesis. The results from this study identify an essential role of the GATA4 protein in the ovarian morphogenetic program. We show that in contrast to the sex determination phase, which relies on the GATA4-FOG2 complex, the subsequent regulation of ovarian differentiation is dependent upon GATA4 but not FOG2. The loss of Gata4 expression within the ovary results in impaired granulosa cell proliferation and theca cell recruitment as well as fewer primordial follicles in the ovarian cortex, causing a failure in follicular development. Preantral follicular atresia is observed within the few follicles that develop despite Gata4 deficiency. The depletion of the follicular pool in GATA4 deficient ovary results in the formation of ovarian cysts and sterility.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Factor de Transcripción GATA4/genética , Regulación del Desarrollo de la Expresión Génica , Folículo Ovárico/embriología , Ovario/embriología , Factores de Transcripción/metabolismo , Alelos , Animales , Diferenciación Celular , Proliferación Celular , Femenino , Eliminación de Gen , Hipotálamo/metabolismo , Ratones , Ratones Endogámicos C57BL , Mutación , Quistes Ováricos/metabolismo , Ovario/metabolismo , Hipófisis/metabolismo , Dedos de Zinc
8.
Domest Anim Endocrinol ; 41(4): 185-94, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21820266

RESUMEN

Gap junctions have been implicated in the regulation of cellular metabolism and the coordination of cellular functions during growth and differentiation of organs and tissues, and gap junctions play a major role in direct cell-cell communication. Gap junctional channels and connexin (Cx) proteins have been detected in adult ovaries in several species. Furthermore, it has been shown that several environmental factors, including maternal diet, may affect fetal organ growth and function. To determine whether maternal diet affects expression of Cx26, Cx32, Cx37, and Cx43 in fetal ovaries, sheep were fed a maintenance (M) diet with adequate (A) selenium (Se) or high (H) Se levels from 21 d before breeding to day 132 of pregnancy. From day 50 to 132 of pregnancy (tissue collection day), a portion of the ewes from the ASe and HSe groups was fed a restricted (R; 60% of M) diet. Sections of fetal ovaries were immunostained for the presence of Cxs followed by image analysis. All four Cxs were detected, but the distribution pattern differed. Cx26 was immunolocalized in the oocytes from primordial, primary, secondary, and antral follicles; in granulosa and theca layers of secondary and antral follicles; stroma; and blood vessels. Cx32 was in oocytes, granulosa, and theca cells in a portion of antral follicles; Cx37 was on the borders between oocyte and granulosa/cumulus cells of primordial to antral follicles and in endothelium; and Cx43 was on cellular borders in granulosa and theca layers and between oocyte and granulosa/cumulus cells of primordial to antral follicles. Maternal diet affected Cx26 and Cx43 expression, Cx26 in granulosa layer of antral follicles was decreased (P < 0.01) by HSe in the M and R diets, and Cx43 in granulosa layer of primary and granulosa and theca of antral follicles was increased (P < 0.05) by the M diet with HSe. Thus, Cxs may be differentially involved in regulation of fetal ovarian function in sheep. These data emphasize the importance of maternal diet in fetal growth and development.


Asunto(s)
Conexinas/metabolismo , Dieta/veterinaria , Feto/metabolismo , Uniones Comunicantes/fisiología , Ovario/metabolismo , Ovinos/metabolismo , Alimentación Animal/análisis , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Conexinas/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/fisiología , Fenómenos Fisiologicos Nutricionales Maternos , Ovario/embriología , Embarazo , Selenio/farmacología , Ovinos/embriología
9.
Mol Hum Reprod ; 15(12): 805-19, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19584195

RESUMEN

Prenatal oogenesis produces hundreds of thousands of oocytes, most of which are discarded through apoptosis before birth. Despite this large-scale selection, the survivors do not constitute a perfect population, and the factors at the cellular level that result in apoptosis or survival of any individual oocyte are largely unknown. What then are the selection criteria that determine the size and quality of the ovarian reserve in women? This review focuses on new data at the cellular level, on human prenatal oogenesis, offering clues about the importance of the timing of entry to meiotic prophase I by linking the stages and progress through MPI with the presence or absence of apoptotic markers. The characteristics and responsiveness of cultured human fetal ovarian tissue at different gestational ages to growth factor supplementation and the impact of meiotic abnormalities upon apoptotic markers are discussed. Future work will require the use of a tissue culture model of prenatal oogenesis in order to investigate the fate of individual live oocytes at different stages of development.


Asunto(s)
Muerte Celular/fisiología , Oocitos , Oogénesis/fisiología , Ovario , Animales , Femenino , Feto/anatomía & histología , Feto/fisiología , Humanos , Etiquetado Corte-Fin in Situ , Profase Meiótica I/fisiología , Ratones , Oocitos/citología , Oocitos/fisiología , Ovario/citología , Ovario/embriología , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/metabolismo
10.
Reproduction ; 137(4): 699-707, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19129369

RESUMEN

Sheep were fed a maintenance (M) diet with adequate (A) Se or high (H) Se concentration from 21 days before breeding to day 135 of pregnancy. From day 50 to day 135 of pregnancy (tissue collection day), a portion of the ewes from ASe and HSe groups were fed restricted (R; 60% of M) diet. Fetal ovarian sections were stained for: 1) the presence of proliferating cell nuclear antigen (a marker of proliferating cells) to determine the proportion of proliferating primordial follicles, or the labeling index (LI; percentage of proliferating cells) for primordial, primary, secondary and antral follicles, stromal tissues, and blood vessels; 2) factor VIII (a marker of endothelial cells) or 3) a presence of apoptotic cells/bodies. The number of proliferating primordial follicles and the LI of primordial follicles was decreased by R and/or HSe diets. The LI was similar for theca and granulosa cells, and for secondary or antral follicles, but was greater in secondary and antral than in primordial and primary follicles. R diet and/or Se affected the LI in all follicle types, in stromal tissues and blood vessels. A dense network of blood vessels was detected in the areas containing secondary to antral follicles, medulla, and hilus, but areas containing primordial follicles were poorly vascularized. The number of apoptotic cells was minimal. These results demonstrate that nutrient restriction and/or Se level in the maternal diet affected cellular proliferation in follicles, blood vessels, and stromal tissues in fetal ovaries. Thus, plane of nutrition and Se in the maternal diet may impact fetal ovarian development and function.


Asunto(s)
Proliferación Celular , Desnutrición/fisiopatología , Fenómenos Fisiologicos Nutricionales Maternos , Ovario/embriología , Selenio/fisiología , Animales , Apoptosis , Restricción Calórica , Femenino , Ovario/irrigación sanguínea , Ovario/fisiología , Embarazo , Ovinos
11.
J Exp Zool A Ecol Genet Physiol ; 307(11): 625-36, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17726668

RESUMEN

Cytochrome P450 aromatase (cyp19) is an enzyme that catalyzes the conversion of androgens to estrogens and may play a role in temperature-dependent sex determination (TSD) of reptiles, amphibians, and fishes. In this study, the ovarian P450 aromatase form (cyp19A1) of pejerrey Odontesthes bonariensis, a teleost with marked TSD, was cloned and its expression profile evaluated during gonadal differentiation at feminizing (17 degrees C, 100% females), mixed-sex producing (24 and 25 degrees C, 73.3 and 26.7% females, respectively), and masculinizing (29 degrees C, 0% females) temperatures. The deduced cyp19A1 amino acid sequence shared high identity (>77.8%) with that from other teleosts but had low identity (<61.8%) with brain forms (cyp19A2), including that of pejerrey itself. The tissue distribution analysis of cyp19A1 mRNA in adult fish revealed high expression in the ovary. Semi-quantitative reverse transcription polymerase chain reaction analysis of the bodies of larvae revealed that cyp19A1 expression increased before the appearance of the first histological signs of ovarian differentiation at the feminizing temperature but remained low at the masculinizing temperature. The expression levels at mixed-sex producing temperatures were bimodal rather than intermediate, showing low and high modal values similar to those at the feminizing and masculinizing temperatures, respectively. The population percentages of high and low expression levels at intermediate temperatures were proportional to the percentage of females and males, respectively, and high levels were first observed at about the time of sex differentiation of females. These results suggest that cyp19A1 is involved in the process of ovarian formation and possibly also in the TSD of pejerrey.


Asunto(s)
Aromatasa/metabolismo , Temperatura Corporal/fisiología , Peces/fisiología , Diferenciación Sexual/fisiología , Secuencia de Aminoácidos , Animales , Aromatasa/genética , Secuencia de Bases , Temperatura Corporal/genética , ADN Complementario/genética , Femenino , Perfilación de la Expresión Génica , Masculino , Datos de Secuencia Molecular , Ovario/embriología , Ovario/metabolismo , ARN Mensajero/metabolismo , Análisis de Secuencia de ADN , Diferenciación Sexual/genética , Razón de Masculinidad , Testículo/embriología , Testículo/metabolismo
12.
Domest Anim Endocrinol ; 31(3): 258-68, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16359839

RESUMEN

The estrogenic and antioxidant effects of the phytoestrogen daidzein (DAI) on germ cell proliferation were evaluated by a chicken ovarian germ-somatic cell coculture model. Ovarian cells were dispersed from 18-day-old embryos, cultured in serum-free McCoy's 5A medium and challenged with DAI alone or in combinations with estrogen receptor antagonist tamoxifen for 48 h. The number of germ cells was counted and the proliferating cells were identified by immunocytochemistry of proliferating cell nuclear antigen (PCNA). The labeling index (LI) was determined for germ cells. Results showed that DAI significantly increased the number of germ cells (P<0.05) and this stimulating effect was inhibited by tamoxifen in a dose-dependent manner. Furthermore, PCNA-LI of germ cells displayed similar changes with the number of germ cells. To estimate the antioxidant action of DAI, ovarian cells were exposed to the reactive oxygen species (ROS)-producing system hypoxanthine/xanthine oxidase (HX/XO). The changes of superoxide dismutase (SOD) activity and glutathione (GSH) level were measured for estimation of the antioxidant status. Ovarian cells were severely damaged by free radicals and this deteriorating effect could be prevented by DAI. Moreover, HX/XO-induced decrease in SOD activity and GSH level was restored by DAI (P<0.05). These results indicated that DAI promoted proliferation of cultured ovarian germ cells by estrogenic action and attenuated ROS-induced toxicity by antioxidant action in embryonic chickens.


Asunto(s)
Antioxidantes/farmacología , Embrión de Pollo/efectos de los fármacos , Isoflavonas/farmacología , Ovario/embriología , Óvulo/efectos de los fármacos , Fitoestrógenos/farmacología , Animales , Procesos de Crecimiento Celular/efectos de los fármacos , Antagonistas de Estrógenos/farmacología , Femenino , Glutatión/metabolismo , Inmunohistoquímica , Microscopía de Contraste de Fase , Ovario/citología , Ovario/efectos de los fármacos , Óvulo/citología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo , Tamoxifeno/farmacología , Xantina Oxidasa/farmacología
13.
Gene Expr Patterns ; 5(5): 655-67, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15939378

RESUMEN

The role of Anti-Müllerian hormone (Amh) during gonad development has been studied extensively in mammals, but is less well understood in other vertebrates. In male mammalian embryos, Sox9 activates expression of Amh, which initiates the regression of the Mullerian ducts and inhibits the expression of aromatase (Cyp19a1), the enzyme that converts androgens to estrogens. To better understand shared features of vertebrate gonadogenesis, we cloned amh cDNA from zebrafish, characterized its genomic structure, mapped it, analyzed conserved syntenies, studied its expression pattern in embryos, larvae, juveniles, and adults, and compared it to the expression patterns of sox9a, sox9b and cyp19a1a. We found that the onset of amh expression occurred while gonads were still undifferentiated and sox9a and cyp19a1a were already expressed. In differentiated gonads of juveniles, amh showed a sexually dimorphic expression pattern. In 31 days post-fertilization juveniles, testes expressed amh and sox9a, but not cyp19a1a, while ovaries expressed cyp19a1a and sox9b, but not amh. In adult testes, amh and sox9a were expressed in presumptive Sertoli cells. In adult ovaries, amh and cyp19a1a were expressed in granulosa cells surrounding the oocytes, and sox9b was expressed in a complementary fashion in the ooplasm of oocytes. The observed expression patterns of amh, sox9a, sox9b, and cyp19a1a in zebrafish correspond to the patterns expected if their regulatory interactions have been conserved with mammals. The finding that zebrafish sox9b and sox8 were not co-expressed with amh in oocytes excludes the possibility that amh expression in zebrafish granulosa cells is directly regulated by either of these two genes.


Asunto(s)
Aromatasa/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Regulación de la Expresión Génica , Glicoproteínas/biosíntesis , Glicoproteínas/genética , Gónadas/embriología , Proteínas HMGB/biosíntesis , Proteínas del Grupo de Alta Movilidad/biosíntesis , Hormonas Testiculares/biosíntesis , Hormonas Testiculares/genética , Factores de Transcripción/biosíntesis , Proteínas de Pez Cebra/biosíntesis , Secuencia de Aminoácidos , Animales , Hormona Antimülleriana , Diferenciación Celular , Mapeo Cromosómico , Clonación Molecular , ADN Complementario/metabolismo , Femenino , Células de la Granulosa/metabolismo , Hibridación in Situ , Masculino , Modelos Genéticos , Datos de Secuencia Molecular , Oocitos/metabolismo , Ovario/embriología , Ovario/metabolismo , Filogenia , Factor de Transcripción SOX9 , Homología de Secuencia de Aminoácido , Testículo/embriología , Testículo/metabolismo , Pez Cebra
14.
Trends Endocrinol Metab ; 13(9): 386-92, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12367820

RESUMEN

Here, we explore the influence of fetal programming and early life exposures on lifelong reproductive health through modification of the hypothalamic-pituitary-gonadal axis. A range of programming issues are considered with examples from the literature demonstrating that environmental or nutritive exposures have a crucial role in reproductive performance, fetal growth, postnatal development and reproduction-related disease risk. We pay particular attention to recent research on associations between indicators of fetal and postnatal growth and the etiology of polycystic ovary syndrome in women. We conclude that the concept of programming can be applied to reproductive development and related health outcomes, and that the complex potential for interactions between parameters controlling fetal development and postnatal exposures invokes a need to adopt a perspective across the life course of an individual.


Asunto(s)
Efectos Tardíos de la Exposición Prenatal , Reproducción , Animales , Dietilestilbestrol/administración & dosificación , Dietilestilbestrol/efectos adversos , Femenino , Hormonas/fisiología , Humanos , Hipotálamo/embriología , Hipotálamo/fisiología , Masculino , Intercambio Materno-Fetal , Neoplasias Ováricas/etiología , Ovario/embriología , Ovario/fisiología , Hipófisis/embriología , Hipófisis/fisiología , Síndrome del Ovario Poliquístico/etiología , Embarazo , Maduración Sexual , Testículo/embriología , Testículo/fisiología
15.
Mech Dev ; 118(1-2): 233-9, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12351194

RESUMEN

Using the digital differential display program of the National Center for Biotechnology Information, we identified a contig of expression sequence tags (ESTs) which were unique to ovary, testis, and egg libraries. The full-length cDNA of this transcript was deduced and further confirmed by reverse transcriptase polymerase chain reaction (RT-PCR). The cDNA encodes a novel protein of 341 amino acids with a nuclear localization signal. The carboxyl-terminus of the protein contains three C2H2 zinc fingers, and the NH(2)-terminus is proline and serine-rich. Based on the conserved zinc finger motifs, we have termed this novel protein as zinc finger protein 393 (ZFP393). Northern blot and RT-PCR analyses revealed that Zfp393 mRNA was exclusively expressed in testis and ovary. The expression sites were further localized by in situ hybridization to step 3-8 spermatids in testis and growing oocytes in ovary. The Zfp393 gene consists of three exons spanning approximately 8 kb on the distal part of mouse chromosome 4. The carboxyl-terminal zinc finger region is highly homologous to several zinc finger-containing proteins, but no proteins were found to share sequence similarity with the NH(2)-terminal region of ZFP393. Genomic database mining and Southern blot analysis indicate that Zfp393 is a single copy gene. We hypothesize that ZFP393 functions as a germ cell-specific transcription factor that plays important roles in spermatid differentiation and oocyte development.


Asunto(s)
Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Dedos de Zinc , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Northern Blotting , Diferenciación Celular , Mapeo Cromosómico , ADN Complementario/metabolismo , Exones , Etiquetas de Secuencia Expresada , Femenino , Perfilación de la Expresión Génica , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Ovario/embriología , Ovario/metabolismo , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Espermatogénesis , Testículo/embriología , Factores de Tiempo , Distribución Tisular , Transcripción Genética
16.
Mech Dev ; 114(1-2): 119-23, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12175496

RESUMEN

In vertebrates, three members of the d4 gene family code for proteins, which are believed to function as transcription factors and involved in regulation of various intracellular processes. One member of the family, ubi-d4/requiem is ubiquitously expressed gene and two other, neuro-d4 and cer-d4, are expressed predominantly in the neural tissues (Nucleic Acids Res. 20 (1992) 5579; Biochim. Biophys. Acta 14 (1992) 172; Mamm. Genome 11 (2000) 72; Mamm. Genome 12 (2001) 862). Typically, d4 proteins show distinct domain organisation with domain 2/3 in the N-terminal, Krüppel-type zinc finger in the central and two adjacent PHD-fingers (d4-domain) in the C-terminal part of the molecule. However, alternative splicing, which is responsible for complex expression patterns of both neurospecific members of the family, generates multiple protein isoforms lacking certain domains (Nucleic Acids Res. 20 (1992) 5579; Genomics 36 (1996) 174; Mamm. Genome 11 (2000) 72; Mamm. Genome 12 (2001) 862). Exact function of d4 proteins is unclear but their involvement in regulation of differentiation and apoptotic cell death has been proposed (J. Biol. Chem. 269 (1994) 29515; Mamm. Genome 11 (2000) 72; Mamm. Genome 12 (2001) 862). Here we identified a single gene, dd4, in the genome of Drosophila melanogaster, the protein product of which could be assigned to the d4 family. Expression of dd4 is regulated during Drosophila development, and is most prominent in syncytial embryos and later in the embryonic nervous and reproductive systems. In flies dd4 mRNA is found in most tissues but the highest level of expression is detected in ovaries.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster/embriología , Expresión Génica , Proteínas de Microtúbulos/biosíntesis , Secuencia de Aminoácidos , Animales , Apoptosis , Femenino , Hibridación in Situ , Masculino , Datos de Secuencia Molecular , Ovario/embriología , Isoformas de Proteínas , Estructura Terciaria de Proteína , ARN Complementario/metabolismo , ARN Mensajero/metabolismo , Homología de Secuencia de Aminoácido , Factores de Tiempo , Distribución Tisular
17.
J Clin Endocrinol Metab ; 87(7): 3417-27, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12107261

RESUMEN

The regulation of germ cell number in the developing ovary is central to female reproduction. Members of the Bcl-2 family of proapoptotic and antiapoptotic proteins have been implicated in this process in rodents. We investigated the expression of Mcl-1, Bcl-2, Bax, and BAD at 13-21 gestational wk in the human fetal ovary and of Mcl-1 in the adult ovary. mRNA expression of Mcl-1 and its short form Mcl-1s, Bcl-2, Bax, and BAD was demonstrated in fetal ovary by RT-PCR. Hybridization array analysis suggested a selective increase in Mcl-1 expression between 14 and 18 wk gestation, which was confirmed by quantitative PCR. There was a corresponding change in the expression of Mcl-1 protein, detected by immunohistochemistry, from germ cells at the periphery of the ovary at 14-16 wk to the largest germ cells, including oocytes within newly formed primordial follicles, at 21 wk. Mcl-1 was also expressed by oocytes of primordial and preantral follicles in the adult. Bax and BAD immunostaining was detected in both somatic and germ cells in the fetal ovary, whereas Bcl-2 was restricted to somatic cells: no changes in expression were observed. Apoptotic cells, detected by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling, were observed in all fetal ovaries but were infrequent. These results confirm that Bcl-2 family members are differentially expressed in several cell types within the developing human ovary. Increased mRNA expression and the changing distribution of Mcl-1 in germ cells as they develop into primordial follicles as well as persistence in the growing oocyte in the adult may indicate an important role for this survival/antiapoptotic factor throughout germ cell development and maturation.


Asunto(s)
Feto/metabolismo , Proteínas de Neoplasias/metabolismo , Oocitos/fisiología , Folículo Ovárico/embriología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , ADN Complementario/genética , Desarrollo Embrionario y Fetal , Femenino , Feto/citología , Humanos , Immunoblotting , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Análisis de Secuencia por Matrices de Oligonucleótidos , Ovario/embriología , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Distribución Tisular , Proteína X Asociada a bcl-2 , Proteína Letal Asociada a bcl
18.
Mol Reprod Dev ; 59(2): 126-32, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11389547

RESUMEN

Melatonin, secreted by the pineal gland, is involved in the regulation of many physiological functions of various species of animals. In the present study, the expression of gene for melatonin Mel(1a) receptor (MelR) was evaluated in the ovary, hypothalamus, and pituitary according to the developmental stages in female mice. Semiquantitative reverse-transcription polymerase chain reaction (RT-PCR) and in situ PCR techniques were applied. According to the developmental stages, gene for MelR was differently expressed on ovary, hypothalamus, and pituitary. MelR gene was first expressed on pituitary prior to the expression in hypothalamus and ovary. Ovarian MelR gene started to express at birth. Unlike hypothalamic expression of MelR gene which was identified after birth, in pituitary, it was expressed at 16 days post coitum. In the ovary, the expression signal of MelR gene was identified on granulosa cells. However, the signal was not detected in the theca cells. It was weak in the primordial and atretic follicles. Taken together, it can be considered that melatonin has a pivotal role in the folliculogenesis.


Asunto(s)
Hipotálamo/metabolismo , Melatonina/metabolismo , Ovario/metabolismo , Hipófisis/metabolismo , Receptores de Superficie Celular/genética , Receptores Citoplasmáticos y Nucleares/genética , Animales , Desarrollo Embrionario y Fetal , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Hipotálamo/citología , Hipotálamo/embriología , Hipotálamo/crecimiento & desarrollo , Hibridación in Situ , Ratones , Neuronas/citología , Neuronas/metabolismo , Especificidad de Órganos , Ovario/citología , Ovario/embriología , Ovario/crecimiento & desarrollo , Hipófisis/citología , Hipófisis/embriología , Hipófisis/crecimiento & desarrollo , Embarazo , Receptores de Superficie Celular/biosíntesis , Receptores Citoplasmáticos y Nucleares/biosíntesis , Receptores de Melatonina , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Mech Dev ; 103(1-2): 107-15, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11335116

RESUMEN

Ovarian development in the domesticated silkmoth, Bombyx mori, is induced by the molting hormone 20-hydroxy-ecdysone (20E) shortly after larval-pupal ecdysis. Studies of the ecdysone response in Drosophila and other insects have shown that 20E exerts its effects initially by the induction of a small number of early genes, including the orphan nuclear receptors HR3, that transduce and amplify the hormone signal. Here we show that the silkmoth orphan receptor BmHR3A acts in the 20E-induced regulatory cascade in the ovary during pupal and pharate adult development in a manner different than that observed in the classical ecdysone regulatory hierarchy in Drosophila salivary glands at the end of the third instar. While other isoforms of BmHR3 are induced as early gene products in the ecdysone response, BmHR3A is induced 2 days after 20E administration in the silkmoth ovary and, thus, behaves as late product. The period of accumulation of BmHR3A in ovarian follicular cells occurs during vitellogenesis and coincides with the period of transcriptional expression of the ESP (egg-specific protein) gene, whose product constitutes a major component of the egg yolk, while it is reciprocal to the period of expression of BmGATAbeta, a gene encoding a regulator of late chorion gene expression. Bandshift experiments demonstrate that BmHR3A binds specifically to RORE (Retinoic acid-related Orphan receptor Response Element)-like sequences in the promoters of both genes, thus suggesting a direct role for BmHR3A in regulating the expression of BmGATAbeta and ESP genes during vitellogenesis. Finally, we show that BmHR3A functions as a constitutive transcriptional activator in a B. mori derived cell line. We propose that BmHR3A may function as a regulator of vitellogenesis in the silkmoth ovary.


Asunto(s)
Bombyx/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ovario/metabolismo , Receptores Citoplasmáticos y Nucleares/biosíntesis , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/fisiología , Secuencia de Aminoácidos , Animales , Northern Blotting , Cloranfenicol O-Acetiltransferasa/metabolismo , ADN Complementario/metabolismo , Femenino , Inmunohistoquímica , Datos de Secuencia Molecular , Ovario/embriología , Biosíntesis de Proteínas , Isoformas de Proteínas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Transcripción Genética
20.
Dev Dyn ; 220(4): 363-76, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11307169

RESUMEN

Two nuclear receptors, Ad4BP/SF-1 and Dax-1, are essential regulators for development and function of the mammalian reproductive system. Similarity in expression sites, such as adrenal glands, gonads, pituitary, and hypothalamus, suggests a functional interaction, and the phenotype similarities were manifested in Ad4BP/SF-1-deficient mice and in cases of natural human mutations of Dax-1. In this study, quantitative reverse transcriptase polymerase chain reaction analyses revealed that expression profiles of Dax-1 in embryonic gonads are different between the two sexes and also from those of Ad4BP/SF-1. Immunohistochemical analyses clarified the spatial and temporal expressions of the Dax-1 protein during development of tissues composing the hypothalamic-pituitary-gonadal axis. During gonadal development, Dax-1 occurred after Ad4BP/SF-1 exhibiting a sexually dimorphic expression pattern at indifferent stages, indicating a possibility of Dax-1 involvement in earliest sex differentiation. When cord formation begins in the testis at embryonic day 12.5 (E12.5), Dax-1 was expressed strongly in Sertoli cells, but its expression level markedly decreased in Sertoli cells and increased in interstitial cells between E13.5 and E17.5. In the female, Dax-1 was strongly expressed in the entire ovarian primordium from E12.5 until E14.5, and then its expression level was decreased and limited to cells near the surface epithelium between E17.5 and postnatal day 0 (P0). During postnatal development of the testis, the variable staining of Dax-1 in Sertoli cells was detected as early as P7 and Dax-1-expressing Leydig cells became rare. In the postnatal ovary, Dax-1 expression was detected in granulosa cells with variable staining intensity, and occasionally in interstitial cells. During pituitary organogenesis, Dax-1 but not Ad4BP/SF-1 was expressed in the dorsal part of Rathke's pouch from E9.5. Later in development after E14.5, the distribution of Dax-1 overlapped with that of Ad4BP/SF-1, being restricted to gonadotropic cells in the anterior pituitary. In the ventromedial hypothalamus (VMH), Dax-1 and Ad4BP/SF-1 were mostly colocalized throughout the embryonic and postnatal development. Thus, the coexpression of Dax-1 and Ad4BP/SF-1 indicates their closely related functions in the development of the reproductive system. Furthermore, we noticed the presence of cells that express Dax-1 but not Ad4BP/SF-1, further indicating additional functions of Dax-1 in an Ad4BP/SF-1-independent molecular mechanism.


Asunto(s)
Proteínas de Unión al ADN/biosíntesis , Gónadas/embriología , Hipotálamo/embriología , Hipófisis/embriología , Receptores de Ácido Retinoico/biosíntesis , Proteínas Represoras , Factores de Transcripción/biosíntesis , Animales , Receptor Nuclear Huérfano DAX-1 , ADN Complementario/metabolismo , Femenino , Factores de Transcripción Fushi Tarazu , Proteínas de Homeodominio , Immunoblotting , Inmunohistoquímica , Células Intersticiales del Testículo/metabolismo , Masculino , Ratones , Ovario/embriología , Receptores Citoplasmáticos y Nucleares , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células de Sertoli/metabolismo , Factores Sexuales , Factor Esteroidogénico 1 , Testículo/embriología , Factores de Tiempo , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA