Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters

Publication year range
1.
Biol Reprod ; 91(5): 111, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25253738

ABSTRACT

Several aspects of equine ovarian physiology are unique among domestic species. Moreover, follicular growth patterns are very similar between horses and humans. This study aimed to characterize, for the first time, global gene expression profiles associated with growth and preovulatory (PO) maturation of equine dominant follicles. Granulosa cells (GCs) and theca interna cells (TCs) were harvested from follicles (n = 5) at different stages of an ovulatory wave in mares corresponding to early dominance (ED; diameter ≥22 mm), late dominance (LD; ≥33 mm) and PO stage (34 h after administration of crude equine gonadotropins at LD stage), and separately analyzed on a horse gene expression microarray, followed by validation using quantitative PCR and immunoblotting/immunohistochemistry. Numbers of differentially expressed transcripts (DETs; ≥2-fold; P < 0.05) during the ED-LD and LD-PO transitions were 546 and 2419 in GCs and 5 and 582 in TCs. The most prominent change in GCs was the down-regulation of transcripts associated with cell division during both ED-LD and LD-PO. In addition, DET sets during LD-PO in GCs were enriched for genes involved in cell communication/adhesion, antioxidation/detoxification, immunity/inflammation, and cholesterol biosynthesis. In contrast, the largest change in TCs during the LD-PO transition was an up-regulation of genes involved in immune activation, with other DET sets mapping to GPCR/cAMP signaling, lipid/amino acid metabolism, and cell proliferation/survival and differentiation. In conclusion, distinct expression profiles were identified between growing and PO follicles and, particularly, between GCs and TCs within each stage. Several DETs were identified that have not been associated with follicle development in other species.


Subject(s)
Gene Expression Profiling , Granulosa Cells/metabolism , Horses , Ovarian Follicle/physiology , Theca Cells/metabolism , Animals , Female , Follicular Phase/genetics , Gene Expression Profiling/veterinary , Horses/physiology , Ovulation/physiology , Transcriptome
2.
Reproduction ; 148(3): 271-83, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24920665

ABSTRACT

Several different miRNAs have been proposed to regulate ovarian follicle function; however, very limited information exists on the spatiotemporal patterns of miRNA expression during follicle development. The objective of this study was to identify, using microarray, miRNA profiles associated with growth and regression of dominant-size follicles in the bovine monovular ovary and to characterize their spatiotemporal distribution during development. The follicles were collected from abattoir ovaries and classified as small (4-8  mm) or large (12-17  mm); the latter were further classified as healthy or atretic based on estradiol and CYP19A1 levels. Six pools of small follicles and individual large healthy (n=6) and large atretic (n=5) follicles were analyzed using Exiqon's miRCURY LNA microRNA Array 6th gen, followed by qPCR validation. A total of 17 and 57 sequences were differentially expressed (greater than or equal to twofold; P<0.05) between large healthy and each of small and large atretic follicles respectively. Bovine miRNAs confirmed to be upregulated in large healthy follicles relative to small follicles (bta-miR-144, bta-miR-202, bta-miR-451, bta-miR-652, and bta-miR-873) were further characterized. Three of these miRNAs (bta-miR-144, bta-miR-202, and bta-miR-873) were also downregulated in large atretic follicles relative to large healthy follicles. Within the follicle, these miRNAs were predominantly expressed in mural granulosa cells. Further, body-wide screening revealed that bta-miR-202, but not other miRNAs, was expressed exclusively in the gonads. Finally, a total of 1359 predicted targets of the five miRNAs enriched in large healthy follicles were identified, which mapped to signaling pathways involved in follicular cell proliferation, steroidogenesis, prevention of premature luteinization, and oocyte maturation.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Granulosa Cells/metabolism , MicroRNAs/metabolism , Ovarian Follicle/metabolism , Animals , Cattle , Female , Gene Expression Profiling , MicroRNAs/genetics , Ovarian Follicle/growth & development
3.
J Cell Sci ; 124(Pt 20): 3484-91, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21984812

ABSTRACT

Important questions in biology have emerged recently concerning the timing of transcription in living cells. Studies on clonal cell lines have shown that transcription is often pulsatile and stochastic, with implications for cellular differentiation. Currently, information regarding transcriptional activity at cellular resolution within a physiological context remains limited. To investigate single-cell transcriptional activity in real-time in living tissue we used bioluminescence imaging of pituitary tissue from transgenic rats in which luciferase gene expression is driven by a pituitary hormone gene promoter. We studied fetal and neonatal pituitary tissue to assess whether dynamic patterns of transcription change during tissue development. We show that gene expression in single cells is highly pulsatile at the time endocrine cells first appear but becomes stabilised as the tissue develops in early neonatal life. This stabilised transcription pattern might depend upon tissue architecture or paracrine signalling, as isolated cells, generated from enzymatic dispersion of the tissue, display pulsatile luminescence. Nascent cells in embryonic tissue also showed coordinated transcription activity over short distances further indicating that cellular context is important for transcription activity. Overall, our data show that cells alter their patterns of gene expression according to their context and developmental stage, with important implications for cellular differentiation.


Subject(s)
Gene Expression Regulation, Developmental , Periodicity , Pituitary Gland/embryology , Pituitary Hormones/metabolism , Animals , Animals, Genetically Modified , Animals, Newborn , Cells, Cultured , Cellular Microenvironment/genetics , Gene Expression Profiling , Luciferases, Firefly/genetics , Luminescent Measurements/methods , Morphogenesis/genetics , Pituitary Gland/metabolism , Pituitary Hormones/genetics , Promoter Regions, Genetic/genetics , Rats
4.
J Cell Sci ; 123(Pt 3): 424-30, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20130141

ABSTRACT

Gene expression in living cells is highly dynamic, but temporal patterns of gene expression in intact tissues are largely unknown. The mammalian pituitary gland comprises several intermingled cell types, organised as interdigitated networks that interact functionally to generate co-ordinated hormone secretion. Live-cell imaging was used to quantify patterns of reporter gene expression in dispersed lactotrophic cells or intact pituitary tissue from bacterial artificial chromosome (BAC) transgenic rats in which a large prolactin genomic fragment directed expression of luciferase or destabilised enhanced green fluorescent protein (d2EGFP). Prolactin promoter activity in transgenic pituitaries varied with time across different regions of the gland. Although amplitude of transcriptional responses differed, all regions of the gland displayed similar overall patterns of reporter gene expression over a 50-hour period, implying overall co-ordination of cellular behaviour. By contrast, enzymatically dispersed pituitary cell cultures showed unsynchronised fluctuations of promoter activity amongst different cells, suggesting that transcriptional patterns were constrained by tissue architecture. Short-term, high resolution, single cell analyses in prolactin-d2EGFP transgenic pituitary slice preparations showed varying transcriptional patterns with little correlation between adjacent cells. Together, these data suggest that pituitary tissue comprises a series of cell ensembles, which individually display a variety of patterns of short-term stochastic behaviour, but together yield long-range and long-term coordinated behaviour.


Subject(s)
Pituitary Gland/metabolism , Prolactin/genetics , Animals , Animals, Genetically Modified , Cells, Cultured , Chromosomes, Artificial, Bacterial/genetics , Fluorescent Antibody Technique , Gene Expression , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , In Vitro Techniques , Luciferases/genetics , Luciferases/metabolism , Male , Microscopy, Fluorescence , Promoter Regions, Genetic/genetics , Rats , Rats, Inbred F344
5.
Reproduction ; 143(1): 21-33, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22016380

ABSTRACT

We investigated the effects of different windows of testosterone propionate (TP) treatment during foetal and neonatal life in female rats to determine whether and when excess androgen exposure would cause disruption of adult reproductive function. Animals were killed prepubertally at d25 and as adults at d90. Plasma samples were taken for hormone analysis and ovaries serial sectioned for morphometric analyses. In prepubertal animals, only foetal+postnatal and late postnatal TP resulted in increased body weights, and an increase in transitory, but reduced antral follicle numbers without affecting total follicle populations. Treatment with TP during both foetal+postnatal life resulted in the development of streak ovaries with activated follicles containing oocytes that only progressed to a small antral (smA) stage and inactive uteri. TP exposure during foetal or late postnatal life had no effect upon adult reproductive function or the total follicle population, although there was a reduction in the primordial follicle pool. In contrast, TP treatment during full postnatal life (d1-25) resulted in anovulation in adults (d90). These animals were heavier, had a greater ovarian stromal compartment, no differences in follicle thecal cell area, but reduced numbers of anti-Mullerian hormone-positive smA follicles when compared with controls. Significantly reduced uterine weights lead reduced follicle oestradiol production. These results support the concept that androgen programming of adult female reproductive function occurs only during specific time windows in foetal and neonatal life with implications for the development of polycystic ovary syndrome in women.


Subject(s)
Ovary/drug effects , Ovary/physiology , Testosterone Propionate/administration & dosage , Testosterone Propionate/adverse effects , Age Factors , Animals , Animals, Newborn , Disease Models, Animal , Female , Fetus/drug effects , Humans , Ovary/abnormalities , Polycystic Ovary Syndrome/etiology , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Wistar , Weight Gain/drug effects
6.
Reproduction ; 144(2): 245-55, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22685252

ABSTRACT

The current study investigated hormonal and ovarian changes during physiological reproductive aging in Sarda ewes. In a first experiment, follicular and corpus luteum dynamics were compared during an induced oestrus cycle in aged (12-14 years) and young adult ewes (4-5 years). Oestrus cycle characteristics did not differ between the two experimental groups. However, follicular function during the follicular phase showed significant alterations in aged ewes, as determined by a lack of dominance effect and by lower mean values of circulating oestradiol (E(2)) and inhibin levels, compared with young adult ewes. In a second experiment, differences in follicle growth, hormonal milieu and oocyte quality in response to exogenous FSH administration were assessed in aged and adult ewes. No differences were recorded in ovarian response to FSH treatment between young adult and aged ewes, as evaluated by ultrasonographic data and circulating concentrations of LH, E(2) and inhibin-A. Although the total number of recovered oocytes was similar in the two age groups, the number of good quality oocytes selected for IVM was significantly lower in aged ewes compared with adult ones. Thereafter, no differences were recorded in cleavage rates, total blastocyst output, embryo developmental kinetic and quality between aged and adult groups. In conclusion, this study demonstrated that reproductive aging in sheep is associated with impaired follicle functionality and an increase in the proportion of oocytes showing morphological abnormalities. However interestingly, oocyte developmental competence in vitro and embryo cryotolerance were not affected by the aging process, when only good quality oocytes were chosen.


Subject(s)
Aging/physiology , Gonadotropins/pharmacology , Ovarian Follicle/drug effects , Age Factors , Aging/blood , Aging/drug effects , Animals , Cryopreservation , Embryo, Mammalian , Estradiol/blood , Female , Fertilization in Vitro , Follicle Stimulating Hormone/administration & dosage , Follicle Stimulating Hormone/pharmacology , In Vitro Oocyte Maturation Techniques , Inhibins/blood , Luteinizing Hormone/blood , Models, Animal , Ovarian Follicle/physiology , Premenopause/blood , Premenopause/physiology , Sheep
7.
Am J Physiol Endocrinol Metab ; 301(2): E288-97, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21521715

ABSTRACT

Secretoneurin (SN) is a functional secretogranin II (SgII)-derived peptide that stimulates luteinizing hormone (LH) production and its release in the goldfish. However, the effects of SN on the pituitary of mammalian species and the underlying mechanisms remain poorly understood. To study SN in mammals, we adopted the mouse LßT2 gonadotropin cell line that has characteristics consistent with normal pituitary gonadotrophs. Using radioimmunoassay and real-time RT-PCR, we demonstrated that static treatment with SN induced a significant increment of LH release and production in LßT2 cells in vitro. We found that GnRH increased cellular SgII mRNA level and total SN-immunoreactive protein release into the culture medium. We also report that SN activated the extracellular signal-regulated kinases (ERK) in either 10-min acute stimulation or 3-h chronic treatment. The SN-induced ERK activation was significantly blocked by pharmacological inhibition of MAPK kinase (MEK) with PD-98059 and protein kinase C (PKC) with bisindolylmaleimide. SN also increased the total cyclic adenosine monophosphate (cAMP) levels similarly to GnRH. However, SN did not activate the GnRH receptor. These data indicate that SN activates the protein kinase A (PKA) and cAMP-induced ERK signaling pathways in the LH-secreting mouse LßT2 pituitary cell line.


Subject(s)
Gonadotrophs/physiology , Luteinizing Hormone, beta Subunit/metabolism , MAP Kinase Signaling System/physiology , Neuropeptides/genetics , Secretogranin II/genetics , Animals , Antibodies/immunology , Antibodies/pharmacology , Chromogranin A/genetics , Chromogranin A/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Gene Expression/drug effects , Gene Expression/physiology , Goldfish , Gonadotrophs/drug effects , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/pharmacology , HEK293 Cells , Humans , Indoles/metabolism , Luteinizing Hormone, beta Subunit/genetics , MAP Kinase Signaling System/drug effects , Maleimides/metabolism , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neuropeptides/immunology , Neuropeptides/pharmacology , Paracrine Communication/drug effects , Paracrine Communication/physiology , Protein Kinase C/antagonists & inhibitors , Secretogranin II/immunology , Secretogranin II/pharmacology
8.
Biol Reprod ; 85(3): 584-93, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21270429

ABSTRACT

While the germ cell-specific RNA binding protein, DAZL, is essential for oocytes to survive meiotic arrest, DAZL heterozygous (het) mice have an increased ovulation rate that is associated with elevated inhibin B and decreased plasma follicle-stimulating hormone (FSH). The relationship between decreased oocyte DAZL expression and enhanced follicular development in het mice was investigated using in vitro follicle cultures and in vivo modulation of endogenous FSH, by treating mice with inhibin and exogenous FSH. In vitro, follicles from het mice are more sensitive to FSH than those of wild-type (wt) mice and can grow in FSH concentrations that are deleterious to wild-type follicles. In vivo, despite no differences between genotypes in follicle population profiles, analysis of granulosa cell areas in antral follicles identified a significantly greater number of antral follicles with increased granulosa cell area in het ovaries. Modulation of FSH in vivo, using decreasing doses of FSH or ovine follicular fluid as a source of inhibin, confirmed the increased responsiveness of het antral follicles to FSH. Significantly more follicles expressing aromatase protein confirmed the earlier maturation of granulosa cells in het mice. In conclusion, it is suggested that DAZL expression represses specific unknown genes that regulate the response of granulosa cells to FSH. If this repression is reduced, as in DAZL het mice, then follicles can grow to the late follicular stage despite declining levels of circulating FSH, thus leading to more follicles ovulating and increased litter size.


Subject(s)
Follicle Stimulating Hormone/physiology , Litter Size , Oocytes/metabolism , Ovarian Follicle/physiology , RNA-Binding Proteins/metabolism , Animals , Aromatase/metabolism , Female , Gonadal Steroid Hormones/metabolism , Immunohistochemistry , Inhibins/metabolism , Luteinizing Hormone/blood , Male , Mice , Ovarian Follicle/cytology , Sheep , Tissue Culture Techniques
9.
Stem Cells ; 28(8): 1368-78, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20506112

ABSTRACT

Primordial germ cells (PGCs) are the embryonic precursors of gametes in the adult organism, and their development, differentiation, and survival are regulated by a combination of growth factors collectively known as the germ cell niche. Although many candidate niche components have been identified through studies on mouse PGCs, the growth factor composition of the human PGC niche has not been studied extensively. Here we report a detailed analysis of the expression of components of the bone morphogenetic protein (BMP) signaling apparatus in the human fetal ovary, from postmigratory PGC proliferation to the onset of primordial follicle formation. We find developmentally regulated and reciprocal patterns of expression of BMP2 and BMP4 and identify germ cells to be the exclusive targets of ovarian BMP signaling. By establishing long-term cultures of human fetal ovaries in which PGCs are retained within their physiological niche, we find that BMP4 negatively regulates postmigratory PGC numbers in the human fetal ovary by promoting PGC apoptosis. Finally, we report expression of both muscle segment homeobox (MSX)1 and MSX2 in the human fetal ovary and reveal a selective upregulation of MSX2 expression in human fetal ovary in response to BMP4, suggesting this gene may act as a downstream effector of BMP-induced apoptosis in the ovary, as in other systems. These data reveal for the first time growth factor regulation of human PGC development in a physiologically relevant context and have significant implications for the development of cultures systems for the in vitro maturation of germ cells, and their derivation from pluripotent stem cells.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Fetus/cytology , Fetus/metabolism , Germ Cells/cytology , Ovary/cytology , Ovary/metabolism , Apoptosis/genetics , Apoptosis/physiology , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein 7/metabolism , Bone Morphogenetic Proteins/genetics , Cell Proliferation , Female , Fluorescent Antibody Technique , Germ Cells/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , MSX1 Transcription Factor/genetics , MSX1 Transcription Factor/metabolism , Ovary/embryology , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction , Tissue Culture Techniques
10.
Reproduction ; 139(2): 395-407, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19900988

ABSTRACT

In humans and domestic mammals, pivotal processes in ovary development, including primordial follicle assembly, occur prenatally. These events are essential for determining fertility in adult life; however, they remain poorly understood at the mechanistic level. In mammals, the SLITs (SLIT1, SLIT2 and SLIT3) and their ROBO (ROBO1, ROBO2, ROBO3/RIG-1 and ROBO4/MAGIC ROBO) receptors regulate neural, leukocyte, vascular smooth muscle cell and endothelial cell migration. In addition, the SLIT/ROBO pathway has functional roles in embryonic development and in the adult ovary by inhibiting cell migration and promoting apoptosis. We therefore characterised follicle formation and investigated the expression and localisation of the ROBO/SLIT pathway in the ovine fetal ovary. Using RT-PCR, we identified SLIT2, SLIT3, ROBO1, ROBO2 and ROBO4 in sheep ovaries harvested across gestation. The real-time quantitative PCR results implied that ROBO2 expression and ROBO4 expression were elevated during the early stages of follicle formation and stayed abundant during primordial follicle maturation (P<0.05). Immunohistochemistry examination demonstrated that ROBO1 was localised to the pre-granulosa cells, while ROBO2, ROBO4 and SLIT2 were expressed in the oocytes of the developing primordial follicle. This indicates that in the fetal ovary, SLIT-ROBO signalling may require an autocrine and paracrine interaction. Furthermore, at the time of increased SLIT-ROBO expression, there was a significant reduction in the number of proliferating oocytes in the developing ovary (P<0.0001). Overall, these results suggest, for the first time, that the SLIT-ROBO pathway is expressed at the time of follicle formation during fetal ovary development.


Subject(s)
Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Ovarian Follicle/metabolism , Ovary/metabolism , Receptors, Immunologic/metabolism , Signal Transduction , Animals , Autocrine Communication , Cell Proliferation , Female , Fertility , Gene Expression Regulation, Developmental , Gestational Age , Glycoproteins/genetics , Immunohistochemistry , Nerve Tissue Proteins/genetics , Ovarian Follicle/embryology , Ovary/embryology , Paracrine Communication , Receptors, Immunologic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sheep , Signal Transduction/genetics , Roundabout Proteins
11.
Mol Endocrinol ; 23(4): 529-38, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19147700

ABSTRACT

We have generated a humanized double-reporter transgenic rat for whole-body in vivo imaging of endocrine gene expression, using the human prolactin (PRL) gene locus as a physiologically important endocrine model system. The approach combines the advantages of bacterial artificial chromosome recombineering to report appropriate regulation of gene expression by distant elements, with double reporter activity for the study of highly dynamic promoter regulation in vivo and ex vivo. We show first that this rat transgenic model allows quantitative in vivo imaging of gene expression in the pituitary gland, allowing the study of pulsatile dynamic activity of the PRL promoter in normal endocrine cells in different physiological states. Using the dual reporters in combination, dramatic and unexpected changes in PRL expression were observed after inflammatory challenge. Expression of PRL was shown by RT-PCR to be driven by activation of the alternative upstream extrapituitary promoter and flow cytometry analysis pointed at diverse immune cells expressing the reporter gene. These studies demonstrate the effective use of this type of model for molecular physiology and illustrate the potential for providing novel insight into human gene expression using a heterologous system.


Subject(s)
Gene Expression , Genes, Reporter/genetics , Prolactin/genetics , Promoter Regions, Genetic , Rats, Transgenic , Animals , Cell Line , Estrogens/metabolism , Female , Humans , Lipopolysaccharides/immunology , Male , Pituitary Gland/cytology , Pituitary Gland/metabolism , Prolactin/metabolism , Rats , Rats, Inbred F344
12.
Dev Biol ; 314(1): 189-99, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18166170

ABSTRACT

Ovarian germ cell survival is dependent upon the formation of primordial follicles, which occurs during fetal life in the human. Activin contributes to germ cell proliferation and survival at this time. SMADs2 and 3 are central elements in the activin signalling pathway and thus indicate sites of activin action. We have investigated the expression and localisation of SMADs2 and 3 in the fetal ovary between 14 and 20 weeks gestation, i.e. preceding and during primordial follicle formation. SMAD3 mRNA expression increased 1.9 fold (P=0.02). SMAD2 and 3 proteins were localised by immunofluorescence to the nuclei of three distinct populations of somatic cells: (a) stromal cells between clusters of germ cells; (b) some somatic cells intermingled with activin beta A-expressing germ cells; (c) pre-granulosa cells surrounding primordial follicles. Germ cells did not express SMAD2 or 3. Activin A increased and follistatin decreased phosphorylation of SMAD2/3 in vitro, and activin increased SMAD2 and decreased KITLG mRNA expression. It therefore appears that somatic cells are the targets for activin signalling in the developing ovary. The effects of activin on germ cells are indirect and include mediation by the kit ligand/c-Kit pathway, rather than being an autocrine germ cell effect.


Subject(s)
Activins/metabolism , Germ Cells/metabolism , Ovary/metabolism , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Stem Cell Factor/biosynthesis , Female , Fetus/metabolism , Follistatin/metabolism , Gene Expression Regulation, Developmental , Gestational Age , Humans , Ovary/cytology , Ovary/embryology , Phosphorylation , Signal Transduction
13.
Reproduction ; 138(6): 921-33, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19713444

ABSTRACT

The control of fecundity is critical in determining mammalian offspring survival. It is regulated principally by the ovulation rate, so that primates and large farm species commonly have a single offspring. Previously, several mutations have been identified in sheep which increase the naturally low ovulation rate; although in some cases homozygous ewes are infertile. In the present study we present a detailed characterization of a novel mutation in growth differentiation factor 9 (GDF9), found in Icelandic Thoka sheep. This mutation is a single base change (A1279C) resulting in a nonconservative amino acid change (S109R) in the C-terminus of the mature GDF9 protein, which is normally expressed in oocytes at all stages of development. Genotyping all animals for which reproductive records were available confirmed this mutation to be associated with increased fecundity in heterozygous ewes and infertility in homozygotes. Analysis of homozygote ovarian morphology and a number of genes normally activated in growing follicles showed that GDF9 was not involved in oocyte activation, but in subsequent development of the follicle. This study highlights the importance of oocyte factors in regulating fertility and provides new information for structural analysis and investigation of the potentially important sites of dimerization or translational modifications required to produce biologically active GDF9. It also provides the basis for the utilization of these animals to enhance sheep production.


Subject(s)
Growth Differentiation Factor 9/genetics , Infertility, Female/genetics , Mutation, Missense , Sheep Diseases/genetics , Sheep/genetics , Amino Acid Sequence , Animals , Base Sequence , Chromosomes, Mammalian , Female , Genetic Predisposition to Disease , Genital Diseases, Female/congenital , Genital Diseases, Female/genetics , Genital Diseases, Female/pathology , Genital Diseases, Female/veterinary , Growth Differentiation Factor 9/metabolism , Homozygote , Infertility, Female/veterinary , Molecular Sequence Data , Mutation, Missense/physiology , Oocytes/metabolism , Organ Specificity/genetics , Polymorphism, Single Nucleotide/physiology , Sheep/physiology , Sheep Diseases/congenital , Sheep Diseases/pathology
14.
Biochem Biophys Res Commun ; 366(1): 36-41, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18053794

ABSTRACT

Both Sp-family factor Specificity protein 1 (Sp1) and LIM homeodomain transcription factor Lhx4 are involved in regulating the development of pituitary gland and nervous system in mammals. Sp1 gene mutation results in death of mouse embryo around day 11 of gestation, and mouse anterior pituitary development is severely hypoplastic after Lhx4 mutation. While Sp1 interacts with the related Lhx3 gene it is unclear whether Sp1 and Lhx4 also interact to regulate their physiological functions. The present study demonstrates that Lhx4 promoter is TATA-less and GC-rich and these sequences are conserved in different species. We have shown using site-directed mutagenesis and the Dual-Glo Luciferase Assay System that within the -515 to +36bp basic activity regions of hLhx4 promoter the GC boxes were important for Sp1 regulation of the hLhx4 promoter. The electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP) experiments confirmed that Sp1 interacted with Lhx4 by directly binding to GC boxes located in Lhx4 promoter. We conclude Sp1 directly regulates Lhx4 gene expression.


Subject(s)
Gene Expression Regulation/physiology , Homeodomain Proteins/metabolism , Kidney/metabolism , Pituitary Gland/metabolism , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism , Animals , Cell Line , Humans , LIM-Homeodomain Proteins , Mice , Signal Transduction/physiology
15.
Mol Hum Reprod ; 14(5): 269-80, 2008 May.
Article in English | MEDLINE | ID: mdl-18436539

ABSTRACT

Epidemiological studies of the impact of environmental chemicals on reproductive health demonstrate consequences of exposure but establishing causative links requires animal models using 'real life' in utero exposures. We aimed to determine whether prolonged, low-dose, exposure of pregnant sheep to a mixture of environmental chemicals affects fetal ovarian development. Exposure of treated ewes (n = 7) to pollutants was maximized by surface application of processed sewage sludge to pasture. Control ewes (n = 10) were reared on pasture treated with inorganic fertilizer. Ovaries and blood were collected from fetuses (n = 15 control and n = 8 treated) on Day 110 of gestation for investigation of fetal endocrinology, ovarian follicle/oocyte numbers and ovarian proteome. Treated fetuses were 14% lighter than controls but fetal ovary weights were unchanged. Prolactin (48% lower) was the only measured hormone significantly affected by treatment. Treatment reduced numbers of growth differentiation factor (GDF9) and induced myeloid leukaemia cell differentiation protein (MCL1) positive oocytes by 25-26% and increased pro-apoptotic BAX by 65% and 42% of protein spots in the treated ovarian proteome were differently expressed compared with controls. Nineteen spots were identified and included proteins involved in gene expression/transcription, protein synthesis, phosphorylation and receptor activity. Fetal exposure to environmental chemicals, via the mother, significantly perturbs fetal ovarian development. If such effects are replicated in humans, premature menopause could be an outcome.


Subject(s)
Environmental Pollutants/pharmacology , Fetal Development/drug effects , Maternal Exposure/adverse effects , Ovary/embryology , Animals , Body Weight/drug effects , Cell Count , Dose-Response Relationship, Drug , Embryo, Mammalian , Environmental Pollutants/administration & dosage , Female , Fetal Weight/drug effects , Maternal-Fetal Exchange/physiology , Mitotic Index , Models, Biological , Oocytes/cytology , Oocytes/drug effects , Organ Size/drug effects , Ovarian Follicle/cytology , Ovarian Follicle/drug effects , Ovary/drug effects , Ovary/physiology , Pregnancy , Sewage/adverse effects , Sheep/embryology
16.
Endocrinology ; 148(8): 3730-9, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17478557

ABSTRACT

Luteolysis in women is associated with an up-regulation of the expression and activity of matrix metalloproteinase-2 (MMP-2), which is inhibited by human chorionic gonadotropin (hCG) during maternal recognition of pregnancy. Because the primary source of MMP-2 is fibroblasts that do not express LH/hCG receptors, we aimed to investigate the regulation of MMP-2. Women with regular cycles having hysterectomy for nonmalignant conditions and women undergoing oocyte retrieval for assisted conception were used in this current study. Novel primary cultures and cocultures of luteinized granulosa cells and fibroblast-like cells in conjunction with human corpora lutea from different stages of the luteal phase were used to investigate the role of activin-A in the corpus luteum. The effect of hCG, activin-A, and follistatin on MMP-2 activity and expression was assessed by gelatin zymography and quantitative RT-PCR in primary cell cultures. Confirmation of signaling pathways involved in the activation of MMP-2 was assessed by immunofluorescence, RT-PCR, and quantitative RT-PCR. In primary cell culture, steroidogenic cells secrete activin-A and its inhibitors, inhibin-A and follistatin. Follistatin expression is up-regulated by hCG (P < 0.05). The fibroblast-like cells producing MMP-2 have the machinery for activin reception, expressing both type I and type II activin receptors and Smad proteins. Activin-A up-regulated both activity and expression of MMP-2 in fibroblast-like cells (P < 0.05). This activity was inhibited in cocultures of luteinized granulosa cells and fibroblast-like cells in the presence of hCG (P < 0.05) or follistatin (P < 0.01). Activin-A is an excellent candidate for an effector molecule in human luteolysis whose paracrine action is inhibited during maternal recognition of pregnancy.


Subject(s)
Activins/metabolism , Corpus Luteum/physiology , Luteal Phase/physiology , Luteolysis/physiology , Activins/antagonists & inhibitors , Cells, Cultured , Chorionic Gonadotropin/pharmacology , Corpus Luteum/cytology , Female , Fibroblasts/cytology , Fibroblasts/physiology , Follistatin/genetics , Follistatin/metabolism , Granulosa Cells/cytology , Granulosa Cells/physiology , Humans , In Vitro Techniques , Inhibins/genetics , Inhibins/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Paracrine Communication/physiology , RNA, Messenger/metabolism , Signal Transduction/physiology , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism
18.
Proc Biol Sci ; 273(1586): 571-7, 2006 Mar 07.
Article in English | MEDLINE | ID: mdl-16537128

ABSTRACT

In societies of cooperative vertebrates, individual differences in contributions to offspring care are commonly substantial. Recent attempts to explain the causes of this variation have focused on correlations between contributions to care and the protein hormone prolactin, or the steroid hormone testosterone. However, such studies have seldom considered the importance of other hormones or controlled for non-hormonal factors that are correlative with both individual hormone levels and contributions to care. Using multivariate statistics, we show that hormone levels explain significant variation in contributions to pup-feeding by male meerkats, even after controlling for non-hormonal effects. However, long-term contributions to pup provisioning were significantly and positively correlated with plasma levels of cortisol rather than prolactin, while plasma levels of testosterone were not related to individual patterns of pup-feeding. Furthermore, a playback experiment that used pup begging calls to increase the feeding rates of male helpers gave rise to parallel increases in plasma cortisol levels, whilst prolactin and testosterone levels remained unchanged. Our findings confirm that hormones can explain significant amounts of variation in contributions to offspring feeding, and that cortisol, not prolactin, is the hormone most strongly associated with pup-feeding in cooperative male meerkats.


Subject(s)
Carnivora/blood , Carnivora/physiology , Feeding Behavior , Hydrocortisone/blood , Animals , Carnivora/psychology , Female , Male , Multivariate Analysis , Paternal Behavior , Prolactin/blood
19.
Reprod Biol ; 6(3): 243-64, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17220951

ABSTRACT

In the current study we investigated whether the developmental status of the two largest follicles (LF1 and LF2) at the time of administration of the first two doses (0 and 12 h) of FSH of a superovulatory treatment influences periovulatory events and embryo yields in sheep. A larger size of LF1 was negatively correlated with embryo recovery (r=-0.608 for 0 h and r=-523 for 12 h, p<0.05), fertilization (r=-0.464 for 12 h, p<0.05) and viability (r=-0.775 for 12 h, p<0.005). Embryo viability rates were also lower when a higher difference between LF1 and LF2 (r=-0.839 for 0 h and r=-0.761 for 12 h, p<0.01) and a smaller size of LF2 (r=0.877 for 0 h and r=0.622 for 12 h, p<0.01) were observed. This indicates the existence of a limit in the follicular size that will be able to give rise a viable embryo. Conversely, a larger size of LF2 at the time of administration of the first two FSH doses was correlated with reduced recovery rates (r=-0.884 for 0 h and r=-0.706 for 12 h, p<0.01), whilst a decreasing size of LF1 and LF2 was correlated with an increased ovulation rate and recovered embryos. The dominance effect appeared to affect the timing of the preovulatory LH surge. Ewes with a higher difference between LF1 and LF2 displayed earlier LH surges (r=-0.420 for 0 h and r=-0.401 for 12 h, p<0.05) which were related to a higher number of non viable embryos (r=-0.777, p<0.05). The fact that superovulatory yields were affected by, both LF1 and LF2 supports the hypothesis of co-dominance effects in sheep.


Subject(s)
Follicle Stimulating Hormone/pharmacology , Ovarian Follicle/drug effects , Ovarian Follicle/physiology , Sheep/physiology , Superovulation , Animals , Cloprostenol/pharmacology , Estrus/drug effects , Female , Fertility Agents, Female/pharmacology , Flurogestone Acetate/pharmacology , Superovulation/drug effects
20.
Mol Endocrinol ; 30(2): 189-200, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26691151

ABSTRACT

The use of bacterial artificial chromosome (BAC) reporter constructs in molecular physiology enables the inclusion of large sections of flanking DNA, likely to contain regulatory elements and enhancers regions that contribute to the transcriptional output of a gene. Using BAC recombineering, we have manipulated a 160-kb human prolactin luciferase (hPRL-Luc) BAC construct and mutated the previously defined proximal estrogen response element (ERE) located -1189 bp relative to the transcription start site, to assess its involvement in the estrogen responsiveness of the entire hPRL locus. We found that GH3 cell lines stably expressing Luc under control of the ERE-mutated hPRL promoter (ERE-Mut) displayed a dramatically reduced transcriptional response to 17ß-estradiol (E2) treatment compared with cells expressing Luc from the wild-type (WT) ERE hPRL-Luc promoter (ERE-WT). The -1189 ERE controls not only the response to E2 treatment but also the acute transcriptional response to TNFα, which was abolished in ERE-Mut cells. ERE-WT cells displayed a biphasic transcriptional response after TNFα treatment, the acute phase of which was blocked after treatment with the estrogen receptor antagonist 4-hydroxy-tamoxifen. Unexpectedly, we show the oscillatory characteristics of hPRL promoter activity in individual living cells were unaffected by disruption of this crucial response element, real-time bioluminescence imaging showed that transcription cycles were maintained, with similar cycle lengths, in ERE-WT and ERE-Mut cells. These data suggest the -1189 ERE is the dominant response element involved in the hPRL transcriptional response to both E2 and TNFα and, crucially, that cycles of hPRL promoter activity are independent of estrogen receptor binding.


Subject(s)
Estrogens/genetics , Estrogens/pharmacology , Prolactin/genetics , Response Elements/genetics , Transcription, Genetic/drug effects , Base Sequence , Cell Line , Estradiol/pharmacology , Humans , Luciferases/metabolism , Molecular Sequence Data , Mutant Proteins/metabolism , Mutation/genetics , Protein Binding/drug effects , Receptors, Estrogen/metabolism , Time Factors , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL