Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Dev Neurobiol ; 77(10): 1144-1160, 2017 10.
Article in English | MEDLINE | ID: mdl-28388016

ABSTRACT

Over the course of a 4-day period of metamorphosis, the Drosophila larval nervous system is remodeled to prepare for adult-specific behaviors. One example is the reorganization of peripheral nerves in the abdomen, where five pairs of abdominal nerves (A4-A8) fuse to form the terminal nerve trunk. This reorganization is associated with selective remodeling of four layers that ensheath each peripheral nerve. The neural lamella (NL), is the first to dismantle; its breakdown is initiated by 6 hours after puparium formation, and is completely removed by the end of the first day. This layer begins to re-appear on the third day of metamorphosis. Perineurial glial (PG) cells situated just underneath the NL, undergo significant proliferation on the first day of metamorphosis, and at that stage contribute to 95% of the glial cell population. Cells of the two inner layers, Sub-Perineurial Glia (SPG) and Wrapping Glia (WG) increase in number on the second half of metamorphosis. Induction of cell death in perineurial glia via the cell death gene reaper and the Diptheria toxin (DT-1) gene, results in abnormal bundling of the peripheral nerves, suggesting that perineurial glial cells play a role in the process. A significant number of animals fail to eclose in both reaper and DT-1 targeted animals, suggesting that disruption of PG also impacts eclosion behavior. The studies will help to establish the groundwork for further work on cellular and molecular processes that underlie the co-ordinated remodeling of glia and the peripheral nerves they ensheath. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 77: 1144-1160, 2017.


Subject(s)
Drosophila/anatomy & histology , Drosophila/growth & development , Animals , Animals, Genetically Modified , Bromodeoxyuridine , Cell Death , Diphtheria Toxin/genetics , Diphtheria Toxin/metabolism , Drosophila/metabolism , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/metabolism , Immunohistochemistry , Larva/anatomy & histology , Larva/growth & development , Larva/metabolism , Metamorphosis, Biological , Microscopy, Confocal , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Neuroglia/cytology , Neuroglia/metabolism , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peripheral Nerves/anatomy & histology , Peripheral Nerves/growth & development , Peripheral Nerves/metabolism
2.
Theriogenology ; 78(9): 1987-96, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23040060

ABSTRACT

Kisspeptides (KiSS) are a recently discovered family of neuropeptides with a central role in regulating the onset of reproductive function in all animals studied to date. We have established biological and physiological evidence for KiSS signaling in the mare. The objective of the current study was to evaluate the physiological and behavioral responses of mares repeatedly given the equine-specific kisspeptpin decapeptide (eKp-10, YRWNSFGLRY-NH(2)) in an effort to shorten the interovulatory period. Administration of eKp-10 (0.5 mg iv every 4 h) to mares beginning on Day 16 postovulation (Group 2) or in estrus (Group 3) did not shorten the mean ± SEM interovulatory interval compared with untreated (Group 1) controls (21.9 ± 1.2, 22 ± 1.2, and 21.5 ± 1.5 days in Groups 1 to 3, respectively; N = 6 per group), nor was there a significant difference in follicle diameter before ovulation among groups, nor number of days treated with eKp-10 for Groups 2 and 3. Mean daily concentrations of FSH, the preovulatory LH surge (timing, mean, and peak concentrations), and mean progesterone concentrations from the newly formed CL were not significantly different among groups. The initiation of treatment was negatively correlated with sexual receptivity (scored 0 to 5: no interest to strong interest) and serum estradiol concentrations, indicating that eKp-10 can significantly disrupt normal sexual receptivity in the estrous mare. This effect on sexual receptivity was short-lived (< 72 h) and the overall change in sexual receptivity score was not significantly different between Groups 2 and 3 (-1.2 ± 0.5 and -1.4 ± 0.4, respectively). However, the day of the cycle that treatment was initiated significant affected the decline in sexual receptivity score, such that the later in the cycle that treatment was initiated, the greater the estimated decrease in sexual receptivity. In conclusion, the linear hypothalamic-pituitary mechanism for KiSS described in other species was not appropriate for the horse and administration of eKp-10 in the seasonally estrous mare may have been outside of the hormone's normal physiological context.


Subject(s)
Estrus/drug effects , Horses/physiology , Kisspeptins/pharmacology , Ovulation Induction/veterinary , Ovulation/drug effects , Animals , Estradiol/blood , Estradiol/metabolism , Female , Follicle Stimulating Hormone , Luteinizing Hormone , Progesterone/blood , Progesterone/metabolism , Sexual Behavior, Animal/drug effects
3.
J Phys Condens Matter ; 24(12): 124102, 2012 Mar 28.
Article in English | MEDLINE | ID: mdl-22394691

ABSTRACT

Water adsorbs and desorbs intact on Pd(111), forming a hydrogen-bonded wetting layer whose structure we examine by low energy electron diffraction (LEED) and He atom scattering (HAS). LEED shows that water forms commensurate (√3 × âˆš3)R30° clusters that aggregate into a partially ordered, approximately (7 × 7) superstructure as the layer completes. HAS indicates that the water layer remains disordered on a local (approximately 10 Å) scale. Based on workfunction measurements and density functional theory simulations we propose that water forms small, flat domains of a commensurate (√3 × âˆš3)R30° water network, separated by disordered domain boundaries containing largely H-down water. This arrangement allows the water layer to adapt its density and relieve the lateral strain associated with adsorbing water in the optimum flat atop adsorption site. We discuss different possibilities for the structure of these domain walls and compare this strain relief mechanism to the highly ordered, large unit cell structures formed on surfaces such as Pt(111).

4.
Soc Reprod Fertil Suppl ; 67: 119-31, 2010.
Article in English | MEDLINE | ID: mdl-21755667

ABSTRACT

The placenta provides the means for nutrient transfer from the mother to the fetus, waste transfer from the fetus to the mother, protection of the fetus from the maternal immune system, and is an active endocrine organ. While many placental functions have been defined and investigated, assessing the function of specific genes expressed by the placenta has been problematic, since classical ablation-replacement methods are not feasible with the placenta. The pregnant sheep has been a long-standing animal model for assessing in vivo physiology during pregnancy, since surgical placement of indwelling catheters into both maternal and fetal vasculature has allowed the assessment of placental nutrient transfer and utilization, as well as placental hormone secretion, under unanesthetized-unstressed steady state sampling conditions. However, in ruminants the lack of well-characterized trophoblast cell lines and the inefficiency of creating transgenic pregnancies in ruminants have inhibited our ability to assess specific gene function. Recently, sheep and cattle primary trophoblast cell lines have been reported, and may further our ability to investigate trophoblast function and transcriptional regulation of genes expressed by the placenta. Furthermore, viral infection of the trophoectoderm layer of hatched blastocysts, as a means for placenta-specific transgenesis, holds considerable potential to assess gene function in the ruminant placenta. This approach has been used successfully to "knockdown" gene expression in the developing sheep conceptus, and has the potential for gain-of-function experiments as well. While this technology is still being developed, it may provide an efficient approach to assess specific gene function in the ruminant placenta.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Placenta/physiology , Animals , Cattle , Cell Line , Female , Pregnancy , Sheep
5.
Anim Reprod Sci ; 103(1-2): 187-92, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-17507186

ABSTRACT

Human chorionic gonadotropin (hCG) was administered to mares in estrus with large, dominant ovarian follicles to initiate follicular and oocyte maturation. Follicular contents were collected at 0, 2, 4 and 6 h after hCG. Epiregulin, amphiregulin and phosphodiesterase (PDE) mRNA contents of granulosa cells (PDE 4D) were determined by reverse transcription and real-time PCR; PDE 3A mRNA content of single oocytes was determined similarly. Copy numbers of mRNA did not increase for PDE 3A or 4D over the time interval studied. Amounts of epiregulin and amphiregulin mRNA were correlated (r=0.98) when log transformed. Epiregulin and amphiregulin mRNA increased (P<0.01) from controls by 4 h after hCG administration, with amphiregulin increasing (P<0.01) by 2 h after hCG administration. Epiregulin and amphiregulin mRNA levels remained elevated (P<0.01) at 6h after hCG. These results indicate that EGF-like growth factors are likely paracrine mediators of the LH signal in the horse.


Subject(s)
Epidermal Growth Factor/biosynthesis , Glycoproteins/biosynthesis , Horses/physiology , Intercellular Signaling Peptides and Proteins/biosynthesis , Oocytes/metabolism , Ovarian Follicle/metabolism , Phosphoric Diester Hydrolases/biosynthesis , Amphiregulin , Animals , Base Sequence , Chorionic Gonadotropin/pharmacology , EGF Family of Proteins , Epidermal Growth Factor/genetics , Epiregulin , Female , Glycoproteins/genetics , Horses/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Molecular Sequence Data , Oocytes/enzymology , Ovarian Follicle/growth & development , Phosphoric Diester Hydrolases/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Random Allocation , Reverse Transcriptase Polymerase Chain Reaction/veterinary
6.
J Neuroendocrinol ; 18(10): 806-9, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16965299

ABSTRACT

Kisspeptins are encoded by the gene KiSS-1 and regulate gonadotrophin-releasing hormone (GnRH) and gonadotrophin secretion in various species, including humans. Here, we quantify gene expression of KiSS-1 in the arcuate nucleus (ARC) across the ovine oestrous cycle and demonstrate an increase in the caudal division of the ARC during the preovulatory period. These data strongly suggest that kisspeptins are involved in the generation of the preovulatory GnRH and luteinising hormone surge.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Estrous Cycle/physiology , Gonadotropin-Releasing Hormone/physiology , Luteinizing Hormone/physiology , Ovulation/physiology , Animals , Estrous Cycle/metabolism , Feedback, Physiological , Female , Gene Expression Regulation , Gonadotropin-Releasing Hormone/metabolism , Luteinizing Hormone/metabolism , Ovulation/metabolism , Progesterone/blood , Sheep
7.
Endocrinology ; 147(9): 4151-9, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16794007

ABSTRACT

Classically, progesterone has been thought to act only through the well-known genomic pathway involving hormone binding to nuclear receptors (nPR) and subsequent modulation of gene expression. However, there is increasing evidence for rapid, nongenomic effects of progesterone in a variety of tissues in mammals, and it seems likely that a membrane PR (mPR) is causing these events. The objective of this study was to isolate and characterize an ovine mPR distinct from the nPR. A cDNA clone was isolated from ovine genomic DNA by PCR. The ovine mPR is a 350-amino acid protein that, based on computer hydrophobicity analysis, possesses seven transmembrane domains and is distinct from the nPR. Message for the ovine mPR was detected in hypothalamus, pituitary, uterus, ovary, and corpus luteum by RT-PCR. In CHO cells that overexpressed a mPR-green fluorescent protein fusion protein, the ovine mPR was localized to the endoplasmic reticulum and not the plasma membrane. Specific binding of 3H-progesterone to membrane fractions was demonstrated in CHO cells that expressed the ovine mPR but not in nontransfected cells. Furthermore, progesterone and 17 alpha-hydroxy-progesterone stimulated intracellular Ca2+ mobilization in CHO cells that expressed ovine mPR in Ca2+-free medium (P < 0.05) but not in CHO cells transfected with empty vector. This rise in intracellular Ca2+ is believed to be from the endoplasmic reticulum as intracellular Ca2+ mobilization is absent when mPR transfected cells are first treated with thapsigargin to deplete Ca2+ stores from the endoplasmic reticulum. Isolation, identification, tissue distribution, cellular localization, steroid binding, and a functional response for a unique intracellular mPR in the sheep are presented.


Subject(s)
Calcium/metabolism , Cell Membrane/chemistry , Cloning, Molecular , Receptors, Progesterone/chemistry , Receptors, Progesterone/genetics , 17-alpha-Hydroxyprogesterone/pharmacology , Amino Acid Sequence , Animals , CHO Cells , Cell Membrane/metabolism , Corpus Luteum/chemistry , Cricetinae , Cricetulus , DNA, Complementary/isolation & purification , Endoplasmic Reticulum/chemistry , Female , Gene Expression , Green Fluorescent Proteins/genetics , Hypothalamus/chemistry , Molecular Sequence Data , Ovary/chemistry , Pituitary Gland/chemistry , Progesterone/metabolism , Progesterone/pharmacology , RNA, Messenger/analysis , Receptors, Progesterone/physiology , Recombinant Fusion Proteins , Reverse Transcriptase Polymerase Chain Reaction , Sheep , Thapsigargin/pharmacology , Transfection , Tritium , Uterus/chemistry
8.
Domest Anim Endocrinol ; 25(1): 121-31, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12963105

ABSTRACT

Steroidogenic factor-1 (SF-1) is a transcription factor originally characterized as a mediator of gene expression in steroidogenic tissues. Studies in SF-1 knockout mice revealed that SF-1 has additional roles at multiple levels of the hypothalamic-pituitary-gonadal axis, including regulation of gene expression in pituitary gonadotropes. Specific binding sites for SF-1 have been demonstrated in several pituitary genes with essential roles in gonadotropin synthesis, including alpha subunit, LHbeta subunit, and GnRH receptor. In studies aimed at identifying physiological factors controlling pituitary expression of SF-1, GnRH has been implicated as a co-regulator of SF-1 and gonadotropin subunit genes. In both rats and ewes, elevated endogenous secretion of GnRH following ovariectomy was associated with increased amounts of SF-1 mRNA in the anterior pituitary gland. Conversely, removal of GnRH input to the pituitary gland by hypothalamic-pituitary disconnection (HPD) in ovariectomized (OVX) ewes reduced SF-1 expression. Despite these changes, however, treatment of OVX ewes with GnRH following HPD only partially restored levels of SF-1 mRNA in the pituitary gland. Therefore, it is possible that regulation of SF-1 gene expression by GnRH during the estrous cycle may involve ovarian hormones or other hypothalamic factors. Additional studies are required to further define the physiological roles of SF-1 in regulation of the hypothalamic-pituitary-gonadal axis in domestic ruminants.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation , Gonadotropins/genetics , Pituitary Gland/metabolism , Sheep , Transcription Factors/genetics , Animals , Binding Sites , DNA-Binding Proteins/physiology , Female , Follicle Stimulating Hormone, beta Subunit/genetics , Fushi Tarazu Transcription Factors , Gene Expression Regulation/drug effects , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/pharmacology , Homeodomain Proteins , Luteinizing Hormone, beta Subunit/genetics , Ovariectomy , Pituitary Gland, Anterior/chemistry , RNA, Messenger/analysis , Rats , Receptors, Cytoplasmic and Nuclear , Steroidogenic Factor 1 , Transcription Factors/physiology
9.
J Endocrinol ; 171(1): 45-55, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11572789

ABSTRACT

Immortalized cell lines have many potential experimental applications including the analysis of molecular mechanisms underlying cell-specific gene expression. We have utilized a recombinant retrovirus encoding the simian virus-40 (SV-40) large T antigen to construct several immortalized cell lines of equine chorionic girdle cell lineage - the progenitor cells that differentiate into the equine chorionic gonadotropin (eCG) producing endometrial cups. Morphologically, the immortalized cell lines appear similar to normal chorionic girdle cells. Derivation of the immortalized cell lines from a chorionic girdle cell lineage was verified by immunological detection of cell-surface antigens specific to equine invasive trophoblasts. The cell lines differed, however, from mature chorionic girdle cells or endometrial cup cells in that they did not produce eCG and did express MHC class I molecules. Thus, these cell lines appear to have been arrested at a stage of development prior to final differentiation into endometrial cup cells. It was also determined that some of these cell lines as well as endometrial cups express the estrogen receptor-related receptor beta gene, but not the glial cell missing gene (GCMa) both of which are expressed in the murine and human placenta. Among these cell lines, three (eCG 50.5, 100.6 and 500.1) express eCG alpha mRNA. Since regulation of eCG alpha subunit gene is largely unknown, we investigated the signal transduction pathways regulating the eCG alpha subunit gene. Both activators of protein kinase A (PKA) and protein kinase C (PKC) induced the expression of eCG alpha subunit expression 3.2 (P<0.05)- and 1.9 (P<0.05)-fold respectively, in the eCG 500.1 cell line. However, activation of these pathways failed to induce eCG beta subunit expression. In conclusion, lines of equine trophoblast cells have been immortalized that display markers characteristic of those with the equine chorionic girdle and endometrial cup cell lineage. A subset of these cells expresses the eCG alpha subunit gene which is responsive to activators of the PKA and PKC signal transduction pathways.


Subject(s)
Antigens, Polyomavirus Transforming , Cell Line, Transformed/metabolism , Glycoprotein Hormones, alpha Subunit/genetics , Gonadotropins, Equine/genetics , Trophoblasts/cytology , Analysis of Variance , Animals , Carcinogenicity Tests , Cell Lineage , Cell Separation/methods , Chorion/cytology , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activation , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation , Horses , Mice , Mice, Nude , Protein Kinase C/metabolism , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology
10.
Biol Reprod ; 65(4): 1150-5, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11566737

ABSTRACT

Oxytocin stimulates a rapid increase in ovine endometrial prostaglandin (PG) F2alpha synthesis. The overall objective of these experiments was to investigate the cellular mechanisms by which oxytocin induces endometrial PGF2alpha synthesis. The objective of experiment 1 was to determine whether G(i) proteins mediate oxytocin-induced PGF2alpha synthesis. Uteri were collected from four ovary-intact ewes on Day 14 postestrus. Caruncular endometrial explants were dissected and subjected to in vitro incubation. Pertussis toxin, an inhibitor of G(i) proteins, had no effect on the ability of oxytocin to induce PGF2alpha synthesis (P > 0.10). The objective of experiment 2 was to determine whether any of the three mitogen-activated protein kinases (MAPKs), extracellular signal regulated protein kinase (ERK1/2), c-Jun N-terminal/stress-activated protein kinase (JNK/SAPK), or p38 MAPK, mediate oxytocin-induced PGF(2alpha) synthesis. Eleven ovary-intact ewes were given an injection of oxytocin (10 IU; i.v.; n = 5) or physiological saline (i.v.; n = 6) on Day 15 postestrus. Uteri were collected 15 min after injection and caruncular endometrium was dissected. Endometrial homogenates were prepared and subjected to Western blotting. Membranes were probed for both total and phosphorylated forms of all three classes of MAPK. All classes of MAPK were detected in ovine endometrium, but oxytocin treatment had no effect on the expression of these proteins (P > 0.10). ERK1/2 was the only phosphorylated MAPK detected and its concentrations were higher in oxytocin-treated ewes (P < 0.01). The objective of experiment 3 was to further investigate the role of ERK1/2 during oxytocin-induced PGF2alpha synthesis. Uteri were collected from four ovary-intact ewes on Day 14 postestrus. Caruncular endometrial explants were dissected and subjected to in vitro incubation. PD98059, a specific inhibitor of ERK1/2 activity, blocked the ability of oxytocin to stimulate PGF(2alpha synthesis in a dose-dependent manner (P < 0.05). These results indicate that the ovine oxytocin receptor is not coupled to G(i) proteins. These results indicate that oxytocin induces phosphorylation of ERK1/2 and that this MAPK appears to mediate oxytocin-induced PGF2alpha synthesis in ovine endometrium.


Subject(s)
Dinoprost/analogs & derivatives , Dinoprost/biosynthesis , Endometrium/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinases/metabolism , Oxytocin/physiology , Sheep/metabolism , Animals , Blotting, Western , Dinoprost/blood , Endometrium/drug effects , Female , GTP-Binding Protein alpha Subunits, Gi-Go/antagonists & inhibitors , MAP Kinase Kinase 4 , Mitogen-Activated Protein Kinase 1/analysis , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases/analysis , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/analysis , Oxytocin/pharmacology , Pertussis Toxin , Phosphorylation , Virulence Factors, Bordetella/pharmacology , p38 Mitogen-Activated Protein Kinases
11.
Tree Physiol ; 21(8): 489-96, 2001 May.
Article in English | MEDLINE | ID: mdl-11359706

ABSTRACT

Many woody species can be propagated from leafy cuttings. However, following rooting, cuttings of Corylus maxima Mill. cv. Purpurea do not always survive the transition from a highly supportive rooting environment (e.g., fog) to a more natural environment where evaporative demand is higher. We found that it is not the supply of water to leaves, but stomatal dysfunction that leads to severe water deficits in the rooted cuttings. Two hours after well-rooted cuttings were transferred from the rooting environment, we were able to relate visible signs of leaf water deficit to high stomatal conductance (g(s)) and low relative water content (R). Small expanding leaves (L3) had unusually high g(s) and lower R than fully expanded leaves (L1). Although high cuticular conductances (g(c)) were occasionally observed in L3, SEM confirmed that increased total leaf conductance (g) was mainly a result of abnormally wide stomatal opening. We measured changes in the ability of stomata to control water loss during rooting by determining stomatal responsiveness to leaf water deficit in detached L1 and L3 harvested from cuttings during the first 75 days after severance from stock plants. Reduced stomatal responsiveness was observed within 7 days of severance, prior to adventitious root formation, and was more pronounced in L3 than in L1. A period of acclimatization after rooting (no leaf wetting, but a vapor pressure deficit of 0.20 kPa) reduced g(s) by 50% in L3 but not in L1, and partially restored stomatal responsiveness in L1 but not in L3. After rooting, the original leaves on the cutting retained substantial capacity for photosynthesis (e.g., in L1, 8 micromol m(-2) s(-1) at a photosynthetic photon flux density of 400 micromol m(-2) s(-1)). The implications of the results for post-rooting acclimatization procedures are discussed.


Subject(s)
Betulaceae/physiology , Plant Leaves/physiology , Trees/physiology , Photosynthesis/physiology , Water/physiology
12.
Mol Endocrinol ; 15(5): 695-703, 2001 May.
Article in English | MEDLINE | ID: mdl-11328852

ABSTRACT

We have used spot fluorescence photobleaching recovery methods to measure the lateral diffusion of GnRH receptor (GnRHR) fused at its C terminus to green fluorescent protein (GFP) after binding of either GnRH agonists or antagonist. Before ligand binding, GnRHR-GFP exhibited fast rates of lateral diffusion (D = 18 +/- 2.8 x 10(-10)cm2 x sec(-1)) and high values for fractional fluorescence recovery (%R) after photobleaching (73 +/- 1%). Increasing concentrations of agonists, GnRH or D-Ala6-GnRH, caused a dose-dependent slowing of receptor lateral diffusion as well as a decreased fraction of mobile receptors. Increasing concentrations of the GnRH antagonist Antide slowed the rate of receptor diffusion but had no effect on the fraction of mobile receptors, which remained high. To determine whether the decrease in %R caused by GnRH agonists was due, in part, to increased receptor self-association, we measured the fluorescence resonance energy transfer efficiency between GnRHR-GFP and yellow fluorescent protein-GNRHR: There was no energy transfer between GnRHR on untreated cells. Treatment of cells with GnRH agonists led to a concentration-dependent increase in the energy transfer between GnRH receptors to a maximum value of 16 +/- 1%. There was no significant energy transfer between GnRH receptors on cells treated with Antide, even at a concentration of 100 nM. These data provide direct evidence that, before binding of ligand, GnRHR exists as an isolated receptor and that binding of GnRH agonists, but not antagonist, leads to formation of large complexes that exhibit slow diffusion and contain receptors that are self-associated.


Subject(s)
Gonadotropin-Releasing Hormone/pharmacology , Hormone Antagonists/pharmacology , Oligopeptides/pharmacology , Receptors, LHRH/physiology , Animals , Bacterial Proteins/chemistry , CHO Cells , Cricetinae , Cyclic AMP/biosynthesis , Cyclic AMP/metabolism , Diffusion , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/metabolism , Green Fluorescent Proteins , Hormone Antagonists/metabolism , Kinetics , Luminescent Proteins/chemistry , Oligopeptides/metabolism , Receptors, LHRH/agonists , Receptors, LHRH/antagonists & inhibitors , Recombinant Fusion Proteins/chemistry , Signal Transduction/physiology , Spectrometry, Fluorescence
13.
Biol Reprod ; 64(3): 898-903, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11207206

ABSTRACT

Rates of internalization of the murine GnRH receptor fused via its C-terminus to green fluorescent protein (GnRH-R-GFP) were examined in Chinese hamster ovary cells (CHO cells) and compared to those of native murine GnRH-R in a clonal murine gonadotroph cell line (LbetaT2 cells). The resulting rates of internalization of murine receptors were then compared with those of sheep GnRH-R in ovine gonadotrophs. Cells were incubated with radioiodinated [D-Ala6]GnRH on ice for 4 h to allow binding of the ligand to GnRH-R, then cells were warmed to 37 degrees C to permit internalization. Surface-bound radioligand began to decrease as soon as the cells were warmed and had decreased significantly within 20 min. A steady-state level of surface-bound radioligand was achieved after 60 min in both CHO cells and LbetaT2 cells (38% and 41%, respectively, of initial value; P < 0.05). Internalization of radioligand began immediately after warming the cells to 37 degrees C, and a significant proportion of surface ligand had been internalized by 20 min. A steady-state maximum of internalization was reached after 60 min in both CHO cells and LbetaT2 cells (29% and 28%, respectively, of total cell-associated ligand; P < 0.05). Changes in surface-bound radioligand and internalized radioligand in sheep pituitary cells were similar to those in CHO cells and LbetaT2 cells, but the amount of radioligand internalized after 60 min (40% of total cell-associated ligand) was 1.4 times higher than in CHO cells and LbetaT2 cells (P < 0.05). In a separate experiment, the effect of estradiol on the rate of internalization of GnRH-R in ovine pituitary cells was examined. Although treatment of ovine pituitary cells with estradiol approximately doubled the number of GnRH receptors, it did not alter either the rate or extent of receptor internalization. These results show that rates of internalization of recombinant murine GnRH-R-GFP in CHO cells and native murine and ovine GnRH-R in LbetaT2 cells and in sheep pituitary cells, respectively, are similar, but amounts of ovine GnRH-R internalized are greater than those for murine GnRH-R. Further, the rate of internalization of occupied receptor is similar in gonadotroph and nongonadotroph cells, and the addition of GFP to the C-terminus of the murine GnRH-R does not alter the rate of internalization.


Subject(s)
Gonadotropin-Releasing Hormone/analogs & derivatives , Receptors, LHRH/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Estradiol/pharmacology , Female , Gonadotropin-Releasing Hormone/metabolism , Green Fluorescent Proteins , Iodine Radioisotopes , Kinetics , Luminescent Proteins/metabolism , Mice , Pituitary Gland, Anterior/drug effects , Pituitary Gland, Anterior/metabolism , Radioligand Assay/methods , Recombinant Fusion Proteins/metabolism , Sheep , Species Specificity
14.
Biol Reprod ; 62(6): 1479-85, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10819747

ABSTRACT

Abnormal follicular and oocyte growth in ovaries of sheep homozygous (II) for the Inverdale gene, FecX(I), suggest that this gene may influence a fundamental event in initiation of folliculogenesis, with two copies of the gene inhibiting growth at the primordial/primary stage. In addition, striking similarities in ovarian morphology between mice deficient in growth and differentiation factor-9 (GDF-9) and II sheep suggest a relationship between the FecX(I) gene and GDF-9 function in the ovary. Therefore, it was hypothesized that GDF-9 mRNA expression would be inhibited in ovaries of II fetal sheep. To test this hypothesis, in situ hybridization was used to characterize GDF-9 mRNA expression in ovaries of homozygous (II), heterozygous (I+), and control (++) fetal sheep at Day 135 of gestation. GDF-9 mRNA expression was localized exclusively to oocytes from the type 1 follicle stage onward in all genotypes and is the first demonstration of GDF-9 mRNA expression in ovaries of fetal sheep. In addition, GDF-9 mRNA expression was detected in oocytes of abnormal type 2 follicles in the ovaries of II sheep. Thus, it does not appear that inhibition of GDF-9 gene expression is the mechanism of action whereby the FecX(I) gene exerts its influence. However, the possibility of translation at specific stages of follicular development cannot presently be ruled out. In addition, the FecX(I) gene may be involved, either directly or indirectly, in regulating expression of receptors for GDF-9. At present, however, neither the FecX(I) gene product nor the GDF-9 receptor has been isolated or characterized.


Subject(s)
Growth Substances/genetics , Infertility, Female/veterinary , Intercellular Signaling Peptides and Proteins , Ovary/embryology , Ovulation/genetics , Sheep/genetics , Animals , Bone Morphogenetic Protein 15 , Female , Follistatin , Gene Expression , Glycoproteins/genetics , Growth Differentiation Factor 9 , Growth Substances/physiology , Heterozygote , Homozygote , In Situ Hybridization , Infertility, Female/genetics , Ovary/metabolism , Proto-Oncogene Proteins c-kit/genetics , RNA, Messenger/analysis , X Chromosome
15.
Endocrinology ; 141(3): 1001-10, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10698176

ABSTRACT

Although the ability of estradiol to enhance pituitary sensitivity to GnRH is established, the underlying mechanism(s) remain undefined. Herein, we find that approximately 9,100 bp of 5' flanking region from the ovine GnRH receptor (oGnRHR) gene is devoid of transcriptional activity in gonadotrope-derived cell lines and is not responsive to either estradiol or GnRH. In stark contrast, this same 9,100 bp promoter fragment directed tissue-specific expression of luciferase in multiple lines of transgenic mice. To test for hormonal regulation of the 9,100-bp promoter, ovariectomized transgenic females were treated with a GnRH antiserum alone or in combination with estradiol. Treatment with antiserum alone reduced pituitary expression of luciferase by 80%. Pituitary expression of luciferase in animals receiving both antiserum and estradiol was approximately 50-fold higher than animals receiving antiserum alone. The estradiol response of the -9,100-bp promoter was equally demonstrable in males. In addition, a GnRH analog (D-Ala-6-GnRH) that does not cross-react with the GnRH antiserum restored pituitary expression of luciferase in males passively immunized against GnRH to levels not different from castrate controls. Finally, treatment with both estradiol and D-Ala-6-GnRH increased pituitary expression of luciferase to a level greater than the sum of the individual treatments suggesting synergistic activation of the transgene by these two hormones. Thus, despite the complete absence of transcriptional activity and hormonal responsiveness in vitro, 9,100 bp of proximal promoter from the oGnRHR gene is capable of directing tissue-specific expression and is robustly responsive to both GnRH and estradiol in transgenic mice. To begin to refine the functional boundaries of the critical cis-acting elements, we next constructed transgenic mice harboring a transgene consisting of 2,700 bp of 5' flanking region from the oGnRHR gene fused to luciferase. As with the -9,100 bp promoter, expression of luciferase in the -2,700 lines was primarily confined to the pituitary gland, brain and testes. Furthermore, the passive immunization-hormonal replacement paradigms described above revealed both GnRH and estradiol responsiveness of the -2,700-bp promoter. Thus, 2,700 bp of proximal promoter from the oGnRHR gene is sufficient for tissue-specific expression as well as GnRH and estradiol responsiveness. Given the inability to recapitulate estradiol regulation of GnRHR gene expression in vitro, transgenic mice may represent one of the few viable avenues for ultimately defining the molecular mechanisms underlying estradiol regulation of GnRHR gene expression.


Subject(s)
Estradiol/pharmacology , Gene Expression Regulation/drug effects , Gonadotropin-Releasing Hormone/pharmacology , Receptors, LHRH/genetics , Animals , Cells, Cultured , DNA/genetics , Female , Genetic Vectors , Luciferases/genetics , Mice , Mice, Transgenic , Ovariectomy , Plasmids/genetics , Receptors, LHRH/biosynthesis , Recombinant Fusion Proteins/genetics , Sheep , Tissue Distribution , Transfection
17.
Mol Endocrinol ; 13(4): 566-77, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10194763

ABSTRACT

Homologous regulation of GnRH receptor (GnRHR) gene expression is an established mechanism for controlling the sensitivity of gonadotropes to GnRH. We have found that expression of the GnRHR gene in the gonadotrope-derived alpha T3-1 cell line is mediated by a tripartite enhancer that includes a consensus activator protein-1 (AP-1) element, a binding site for SF-1 (steroidogenic factor-1), and an element we have termed GRAS (GnRHR-activating sequence). Further, in transgenic mice, approximately 1900 b.p. of the murine GnRHR gene promoter are sufficient for tissue-specific expression and GnRH responsiveness. The present studies were designed to further delineate the molecular mechanisms underlying GnRH regulation of GnRHR gene expression. Vectors containing 600 bp of the murine GnRHR gene promoter linked to luciferase (LUC) were transiently transfected into alpha T3-1 cells and exposed to treatments for 4 or 6 h. A GnRH-induced, dose-dependent increase in LUC expression of the -600 promoter was observed with maximal induction of LUC noted at 100 nM GnRH. We next tested the ability of GnRH to stimulate expression of vectors containing mutations in each of the components of the tripartite enhancer. GnRH responsiveness was lost in vectors containing mutations in AP-1. Gel mobility shift data revealed binding of fos/jun family members to the AP-1 element of the murine GnRHR promoter. Treatment with GnRH or phorbol-12-myristate-13-acetate (PMA) (100 nM), but not forskolin (10 microM), increased LUC expression, which was blocked by the protein kinase C (PKC) inhibitor, GF109203X (100 nM), and PKC down-regulation (10 nM PMA for 20 h). In addition, a specific MEK1/MEK2 inhibitor, PD98059 (60 microM), reduced the GnRH and PMA responses whereas the L-type voltage-gated calcium channel agonist, +/- BayK 8644 (5 microM), and antagonist, nimodipine (250 nM), had no effect on GnRH responsiveness. Furthermore, treatment of alpha T3-1 cells with 100 nM GnRH stimulated phosphorylation of both p42 and p44 forms of extracellular signal-regulated kinase (ERK), which was completely blocked with 60 microM PD98059. We suggest that GnRH regulation of the GnRHR gene is partially mediated by an ERK-dependent activation of a canonical AP-1 site located in the proximal promoter of the GnRHR gene.


Subject(s)
Protein Kinase C/metabolism , Receptors, LHRH/genetics , Response Elements/physiology , Transcription Factor AP-1/metabolism , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Animals , Binding Sites , Calcium/metabolism , Calcium Channel Agonists/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels/drug effects , Calcium Channels/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Genes, fos , Genes, jun , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/pharmacology , Mice , Nimodipine/pharmacology , Promoter Regions, Genetic , Receptors, LHRH/drug effects , Receptors, LHRH/metabolism , Response Elements/drug effects , Steroidogenic Factor 1 , Transcription Factors/metabolism
18.
Biochem Biophys Res Commun ; 255(2): 382-5, 1999 Feb 16.
Article in English | MEDLINE | ID: mdl-10049717

ABSTRACT

The possibility that LH receptors exist as isolated molecules when unbound and aggregate upon binding gonadotropins has previously been untestable in viable cells for want of a suitable nonhormone probe. We have now expressed in CHO cells an intrinsically-fluorescent LH receptor involving enhanced green fluorescent protein (GFP) fused to the C-terminus of the rat LH receptor (rLHR-GFP). More than half of these receptors (54 +/- 4%) are located on the plasma membrane and are functional: cAMP levels increase 3-5 fold in response to 10 nM LH or hCG. In fluorescence photobleaching recovery studies at 37 degrees C, 54 +/- 13% of unoccupied rLHR-GFP were laterally mobile with a diffusion coefficient D of 16 +/- 3.5 x 10(-10)cm2sec-1. Introduction of 10 nM LH for 1 h slowed receptor lateral diffusion to 6.6 +/- 1.3 x 10(-10)cm2sec-1 and reduced fluorescence recovery after photobleaching to 27 +/- 1%. Following treatment with 1 nM hCG, rLHR-GFP were laterally immobile and were distributed into small fluorescent patches over the cell surface. Thus, unoccupied rLHR-GFP receptors apparently exist as dispersed plasma membrane proteins with comparatively fast lateral diffusion. Interaction of receptors with LH or hCG caused clustering of rLHR-GFP receptors, significantly restricting lateral diffusion.


Subject(s)
Luminescent Proteins/metabolism , Receptor Aggregation , Receptors, LH/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Animals , CHO Cells , Cell Membrane/chemistry , Cell Membrane/metabolism , Chorionic Gonadotropin/metabolism , Cricetinae , Cyclic AMP/biosynthesis , Green Fluorescent Proteins , Humans , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Microscopy, Confocal , Microscopy, Fluorescence , Receptors, LH/biosynthesis , Receptors, LH/genetics , Recombinant Fusion Proteins/biosynthesis
19.
Endocrinology ; 140(4): 1949-52, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10098536

ABSTRACT

Expression of the FSHbeta subunit and GnRH receptor (GnRHR) genes in gonadotropes is stimulated by activin. We sought to identify the cis-acting element(s) in the murine GnRHR gene promoter which confer activin responsiveness. We established that 600 bp of 5'flanking sequence from the murine GnRHR gene were sufficient to confer activin responsiveness in the gonadotrope-derived alphaT3-1 cell line. Since alphaT3-1 cells, like gonadotropes, secrete activin, we examined the ability of follistatin, an activin binding protein, to block the activin response. Increasing concentrations of follistatin from 0 to 100 ng/ml resulted in a dose dependent decrease in activity of the -600 promoter. Contained within this region are three elements important for expression in alphaT3-1 cells: a Steroidogenic Factor-1 binding site (SF-1), an Activator Protein-1(AP-1) element, and an element termed the GnRH receptor activating sequence or GRAS. A block mutation of GRAS inhibited the ability of the promoter to respond to follistatin. A more refined analysis using a series of two-bp mutations which scan GRAS and flanking sequence revealed exact convergence of GRAS with activin/follistatin responsiveness. Finally, a construct consisting of 3 copies of GRAS placed upstream of a heterologous minimal promoter (3xGRAS-PRL-LUC) was responsive to both activin stimulation and follistatin inhibition in alphaT3-1 cells. Thus, autocrine/paracrine stimulation of gonadotropes by activin illustrates a unique mechanism for cell-specific gene expression.


Subject(s)
Gene Expression , Inhibins/pharmacology , Pituitary Gland, Anterior/metabolism , Receptors, LHRH/genetics , Response Elements , Activins , Animals , Cell Line , Follistatin , Glycoproteins/administration & dosage , Glycoproteins/pharmacology , Humans , Mice , Mutagenesis , Prolactin/genetics , Promoter Regions, Genetic , Recombinant Proteins/pharmacology , Transfection
20.
Endocrinology ; 140(2): 950-7, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9927328

ABSTRACT

The GnRH receptor (GnRHR) is a G protein-coupled receptor expressed by gonadotropes in the anterior pituitary gland. In the past several years, much has been learned about the structure-function relationships that exist in this receptor with regard to ligand binding and signal transduction. However, the lack of specific antibodies has precluded any analyses of the behavior of the unbound form of this receptor. We have constructed a functional GnRHR in which enhanced green fluorescent protein is fused to the carboxyl-terminus of the murine GnRHR. This fusion receptor was expressed diffusely throughout the cell, with approximately 38% of the fusion receptors colocalized with a plasma membrane marker in the gonadotrope-derived alphaT3 cell line, and approximately 82% of the fusion receptors colocalized with a membrane marker in Chinese hamster ovary cells. Furthermore, the fusion receptor displayed a Kd of 0.8 nM for iodinated des-Gly10,D-Ala-6-GnRH N-ethyl amide in Chinese hamster ovary cells, which was similar to the Kd of the native GnRHR expressed in alphaT3 cells. The surface mobility of the fusion protein was examined by fluorescence photobleaching recovery methods. In the unbound state the majority of the receptors were laterally mobile and displayed a lateral diffusion rate of 1.2-1.6 x 10(-9) cm2/sec. Binding of GnRH reduced the rate of lateral diffusion over 3-fold and reduced the fraction of mobile receptors from approximately 76-91% to 44-61%. Like GnRH, the competitive GnRH antagonist antide slowed the rate of receptor diffusion approximately 3-fold. In contrast to GnRH, antide had no effect on the fraction of mobile receptors. Thus, an intrinsically fluorescent GnRHR is trafficked to the plasma membrane of mammalian cells, is capable of ligand binding and signal transduction, and allows direct observation of the GnRHR in the nonligand-bound state. Furthermore, fluorescence photobleaching recovery analysis of the GnRHR-green fluorescent protein fusion reveals fundamental differences in the membrane dynamics of the GnRHR induced by the binding of an agonist vs. that induced by the binding of an antagonist.


Subject(s)
Receptors, LHRH/physiology , Animals , Binding, Competitive , CHO Cells , COS Cells , Cell Membrane/metabolism , Cricetinae , Diffusion , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/metabolism , Green Fluorescent Proteins , Indicators and Reagents , Ligands , Luminescent Proteins/genetics , Mice , Receptors, LHRH/genetics , Receptors, LHRH/metabolism , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...