Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
Add more filters











Publication year range
1.
Endocrinology ; 148(7): 3299-306, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17395696

ABSTRACT

Cultures of endogenous GnRH neurons and the GT1 GnRH neuronal cell line release GnRH in pulses (intrinsic pulsatile release) with an interpulse frequency similar to that seen in castrated animals. In both GT1 cells and transgenic rats, lowering cAMP levels by expression of a phosphodiesterase decreased the frequency of intrinsic GnRH pulsatility. We asked whether the cyclic nucleotide-gated cation (CNG) channels expressed in GT1 cells participated in cAMP modulation of intrinsic GnRH pulsatility. Because expression of the CNGA2 subunit is essential for formation of functional CNG channels, we developed an adenovirus (Ad) vector expressing a short interference RNA (siRNA) to the CNGA2 subunit (Ad-CNG-siRNA) or as an infection control, to the coding region of luciferase (Ad-Luc-siRNA). Infection with the Ad-CNG-siRNA of COS cells transfected with a CNGA2 expression vector significantly inhibited CNGA2 protein levels by 74% by Western blot. Infection of GT1-1 cells with Ad-CNG-siRNA resulted in a 68% decrease in the levels of CNGA2 mRNA, a 44% decrease in protein levels, and a clear decrease in immunostaining with an antibody to CNGA2. Infection of GT1-1 cells with Ad-CNG-siRNA decreased spontaneous Ca2+ oscillations compared with Ad-Luc-siRNA-infected or uninfected cells by 71%. Furthermore infection with Ad-CNG-siRNA resulted in a 2-fold increase in the interpulse interval in GnRH secretion (49.4+/-9.1 min) compared with uninfected cells (25.9+/-2.5 min) or Ad-Luc-siRNA (29.3+/-2.8 min)-infected cells. These data provide the first direct evidence that the CNG channel is a downstream signaling molecule in the regulation of the frequency of intrinsic GnRH pulsatility by cAMP.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Ion Channels/metabolism , Adenoviridae/genetics , Animals , Blotting, Western , COS Cells , Calcium/metabolism , Cell Line , Chlorocebus aethiops , Colforsin/pharmacology , Cyclic AMP/metabolism , Cyclic Nucleotide-Gated Cation Channels , Gene Expression/drug effects , Genetic Vectors/genetics , Humans , Ion Channels/genetics , Ion Channels/physiology , Models, Biological , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , RNA, Small Interfering/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Transfection
2.
Growth Horm IGF Res ; 11 Suppl A: S9-15, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11527095

ABSTRACT

The frequency and amplitude of gonadotropin-releasing hormone (GnRH) pulses are tightly regulated for the maintenance of reproductive cycles. Pulsatile GnRH release was shown to be an intrinsic property of murine GT1 GnRH neurons, and primate placodal GnRH neurons. GT1 neurons show spontaneous action potentials that are associated with Ca2+ oscillations and hormone secretion. Increased cyclic AMP (cAMP) levels in GT1 neurons appear to stimulate GnRH release by activation of cAMP-gated cation (CNG) channels. Activation of the CNG channels correlated with increased neuron excitability and Ca2+ oscillations. Activation of protein kinase A is not necessary for cAMP-induced stimulation of GnRH secretion, but appears to activate negative feedback pathways. Potential negative feedback pathways may decrease cAMP levels by inhibiting adenylyl cyclase V, and activating the phosphodiesterase, PDE4D3. These stimulatory and inhibitory cAMP-signalling pathways appear to regulate the excitability of the GT1 neurons, and may constitute a biological clock timing the pulsatile release of GnRH.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Gonadotropin-Releasing Hormone/metabolism , Phosphoric Diester Hydrolases/metabolism , Signal Transduction , Adenylyl Cyclases/metabolism , Animals , Cyclic Nucleotide-Gated Cation Channels , Humans , Ion Channels/metabolism
3.
Mol Endocrinol ; 14(10): 1536-49, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11043570

ABSTRACT

We asked whether the antiangiogenic action of 16K human PRL (hPRL), in addition to blocking mitogen-induced vascular endothelial cell proliferation, involved activation of programmed cell death. Treatment with recombinant 16K hPRL increased DNA fragmentation in cultured bovine brain capillary endothelial (BBE) and human umbilical vein endothelial (HUVE) cells in a time- and dose-dependent fashion, independent of the serum concentration. The activation of apoptosis by 16K hPRL was specific for endothelial cells, and the activity of the peptide could be inhibited by heat denaturation, trypsin digestion, and immunoneutralization, but not by treatment with the endotoxin blocker, polymyxin-B. 16K hPRL-induced apoptosis was correlated with the rapid activation of caspases 1 and 3 and was blocked by pharmacological inhibition of caspase activity. Caspase activation was followed by inactivation of two caspase substrates, poly(ADP-ribose) polymerase (PARP) and the inhibitor of caspase-activated deoxyribonuclease (DNase) (ICAD). Furthermore, 16K hPRL increased the conversion of Bcl-X to its proapoptotic form, suggesting that the Bcl-2 protein family may also be involved in 16K hPRL-induced apoptosis. These findings support the hypothesis that the antiangiogenic action of 16K hPRL includes the activation of programmed cell death of vascular endothelial cells.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Endothelium, Vascular/cytology , Neovascularization, Physiologic/drug effects , Peptide Fragments/pharmacology , Prolactin/pharmacology , Antibodies, Monoclonal/pharmacology , Cell Division/drug effects , Cell Line , DNA/metabolism , DNA Fragmentation/drug effects , Drug Contamination , Endotoxins/pharmacology , Enzyme Activation/drug effects , Escherichia coli , Female , Hot Temperature , Humans , Molecular Weight , Peptide Fragments/immunology , Prolactin/immunology , Protein Denaturation , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Prolactin/physiology , Receptors, Somatotropin/physiology , Recombinant Proteins/pharmacology , Trypsin/pharmacology
4.
Proc Natl Acad Sci U S A ; 97(4): 1861-6, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10677547

ABSTRACT

We studied the signaling pathways coupling gonadotropin-releasing hormone (GnRH) secretion to elevations in cAMP levels in the GT1 GnRH-secreting neuronal cell line. We hypothesized that increased cAMP could be acting directly by means of cyclic nucleotide-gated (CNG) cation channels or indirectly by means of activation of cAMP-dependent protein kinase (PKA). We showed that GT1 cells express the three CNG subunits present in olfactory neurons (CNG2, -4.3, and -5) and exhibit functional cAMP-gated cation channels. Activation of PKA does not appear to be necessary for the stimulation of GnRH release by increased levels of cAMP. In fact, pharmacological inhibition of PKA activity caused an increase in the basal secretion of GnRH. Consistent with this observation activation PKA inhibited adenylyl cyclase activity, presumably by inhibiting adenylyl cyclase V expressed in the cells. Therefore, the stimulation of GnRH release by elevations in cAMP appears to be the result of depolarization of the neurons initiated by increased cation conductance by cAMP-gated cation channels. Activation of PKA may constitute a negative-feedback mechanisms for lowering cAMP levels. We hypothesize that these mechanisms could result in oscillations in cAMP levels, providing a biochemical basis for timing the pulsatile release of GnRH.


Subject(s)
Cyclic AMP/pharmacology , Gonadotropin-Releasing Hormone/metabolism , Ion Channels/genetics , Signal Transduction , Sulfonamides , Adenylyl Cyclases/metabolism , Animals , Cell Line , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide-Gated Cation Channels , Dopamine/pharmacology , Electrophysiology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Ion Channels/metabolism , Isoquinolines/pharmacology , Mice , Neurons/metabolism , Patch-Clamp Techniques
5.
Neuroendocrinology ; 70(4): 221-7, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10529616

ABSTRACT

Gap junctions (GJs) are transmembrane channels that permit rapid intercellular transit of various small molecules including ions, second messengers and metabolites. GJs promote communication and coordinated activity between coupled neurons, and may help facilitate the synchronous release and pulsatile secretion of neurohormones. A previous study using GnRH-secreting GT1-7 cells reported that connexin 26 was the major GJ subunit present, and that about 20% of the cultured cells engaged in GJ coupling as assayed by fluorescence recovery after photobleaching of 5,6-carboxyfluorescein diacetate (MW 460 D). To reassess GJ connectivity with a more permeant probe, we grew GT1-7 cells to 70% confluency on Matrigel-coated glass coverslips and microinjected Neurobiotin(TM) (MW 322 D) into single cells. Dye was allowed to diffuse for 30 min before cultures were fixed, and subsequently immunostained for Neurobiotin with 3,3'-diaminobenzidine HCl and examined by light microscopy. Dye coupling between 2 or more GT1-7 cells was observed after 75% of all microinjections. Connectivity involved the somata and neurites of an average of 6.6 +/- 2.0 adjoining cells, but in one instance was seen in a group of 32 GT1-7 neighbors. Western blotting and immunofluorescence staining confirmed that connexin 26 was the predominant GJ subunit expressed by GT1-7 cultures. Our results using Neurobiotin suggest these GJ channels may be smaller than anticipated. In addition, functional GJ connectivity between subconfluent GT1-7 cells is more extensive than previously reported, occurring with higher frequency and coupling significantly greater numbers of cultured cells. Since cAMP, IP3, and Ca(2+) are able to pass through GJs and can elicit secretion of GnRH by GT1 cell cultures, GJs may play an important role in the coordination and synchronization of GnRH release.


Subject(s)
Connexins/biosynthesis , Gap Junctions/metabolism , Gonadotropin-Releasing Hormone/metabolism , Neurons/metabolism , Animals , Biotin/analogs & derivatives , Biotin/chemistry , Biotin/pharmacokinetics , Blotting, Western , Cell Line , Connexin 26 , Connexins/analysis , Fluorescent Antibody Technique , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacokinetics , Gap Junctions/chemistry , Hypothalamus/cytology , Isoquinolines/chemistry , Isoquinolines/pharmacokinetics , Molecular Weight , Neurons/chemistry , Neurons/cytology
6.
Mol Endocrinol ; 13(5): 692-704, 1999 May.
Article in English | MEDLINE | ID: mdl-10319320

ABSTRACT

Signaling pathways mediating the antiangiogenic action of 16K human (h)PRL include inhibition of vascular endothelial growth factor (VEGF)-induced activation of the mitogen-activated protein kinases (MAPK). To determine at which step 16K hPRL acts to inhibit VEGF-induced MAPK activation, we assessed more proximal events in the signaling cascade. 16K hPRL treatment blocked VEGF-induced Raf-1 activation as well as its translocation to the plasma membrane. 16K hPRL indirectly increased cAMP levels; however, the blockade of Raf-1 activation was not dependent on the stimulation of cAMP-dependent protein kinase (PKA), but rather on the inhibition of the GTP-bound Ras. The VEGF-induced tyrosine phosphorylation of the VEGF receptor, Flk-1, and its association with the Shc/Grb2/Ras-GAP (guanosine triphosphatase-activating protein) complex were unaffected by 16K hPRL treatment. In contrast, 16K hPRL prevented the VEGF-induced phosphorylation and dissociation of Sos from Grb2 at 5 min, consistent with inhibition by 16K hPRL of the MEK/MAPK feedback on Sos. The inhibition of Ras activation was paralleled by the increased phosphorylation of 120 kDa proteins comigrating with Ras-GAP. Taken together, these findings show that 16K hPRL inhibits the VEGF-induced Ras activation; this antagonism represents a novel and potentially important mechanism for the control of angiogenesis.


Subject(s)
Endothelial Growth Factors/metabolism , Endothelium, Vascular/metabolism , Genes, ras , Lymphokines/metabolism , Prolactin/metabolism , Biological Transport , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/drug effects , Enzyme Activation/drug effects , GRB10 Adaptor Protein , GTPase-Activating Proteins , Humans , Lymphokines/pharmacology , Membrane Proteins/metabolism , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Prolactin/pharmacology , Proteins/metabolism , Proto-Oncogene Proteins c-raf/drug effects , Proto-Oncogene Proteins c-raf/metabolism , Receptor Protein-Tyrosine Kinases/drug effects , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/drug effects , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Signal Transduction , Son of Sevenless Proteins , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , ras GTPase-Activating Proteins , ras Proteins/metabolism
7.
Proc Natl Acad Sci U S A ; 96(4): 1246-51, 1999 Feb 16.
Article in English | MEDLINE | ID: mdl-9990009

ABSTRACT

Angiogenesis, the process of development of a new microvasculature, is regulated by a balance of positive and negative factors. We show both in vivo and in vitro that the members of the human prolactin/growth hormone family, i.e., human prolactin, human growth hormone, human placental lactogen, and human growth hormone variant are angiogenic whereas their respective 16-kDa N-terminal fragments are antiangiogenic. The opposite actions are regulated in part via activation or inhibition of mitogen-activated protein kinase signaling pathway. In addition, the N-terminal fragments stimulate expression of type 1 plasminogen activator inhibitor whereas the intact molecules have no effect, an observation consistent with the fragments acting via separate receptors. The concept that a single molecule encodes both angiogenic and antiangiogenic peptides represents an efficient model for regulating the balance of positive and negative factors controlling angiogenesis. This hypothesis has potential physiological importance for the control of the vascular connection between the fetal and maternal circulations in the placenta, where human prolactin, human placental lactogen, and human growth hormone variant are expressed.


Subject(s)
Endothelium, Vascular/physiology , Human Growth Hormone/physiology , Neovascularization, Physiologic/physiology , Peptide Fragments/pharmacology , Plasminogen Activator Inhibitor 1/biosynthesis , Prolactin/physiology , Allantois/blood supply , Animals , Brain , Capillaries , Cattle , Cell Division/drug effects , Cells, Cultured , Chick Embryo , Chorion/blood supply , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Genetic Variation , Human Growth Hormone/chemistry , Human Growth Hormone/pharmacology , Humans , Kinetics , Neovascularization, Physiologic/drug effects , Placental Lactogen/chemistry , Placental Lactogen/pharmacology , Placental Lactogen/physiology , Plasminogen Activator Inhibitor 1/analysis , Prolactin/chemistry , Prolactin/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology
8.
Endocrinology ; 139(9): 3696-703, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9724020

ABSTRACT

The N-terminal fragment of PRL (16K PRL) is an antiangiogenic factor that, in vitro, inhibits several components of angiogenesis including basic fibroblast growth factor (bFGF)-induced cell division, migration, and organization of capillary endothelial cells. An essential step in the regulation of angiogenesis is the activation of urokinase (urokinase type plasminogen activator, uPA), which in turn activates a cascade of proteases that play essential roles in endothelial cell migration and tissue remodeling. Treatment of bovine capillary endothelial cells (BBEC) with 16K PRL inhibited bFGF-stimulated urokinase activity in BBEC as detected by plasminogen substrate gel assay. 16K PRL did not appear to be acting via an effect on uPA expression because no change in messenger RNA levels were observed. However, protein levels of plasminogen activator inhibitor-1 (PAI-1), a specific inhibitor of urokinase, were increased by 16K PRL independent of the action of bFGF. The 16K PRL-induced increase in PAI-1 protein levels appear to be the result of increased expression of the PAI-1 gene. Increased production of PAI-1 induced by 16K PRL results in the formation of inactive PAI-1/uPA complexes, consistent with the observed decrease in uPA activity.


Subject(s)
Neovascularization, Physiologic/drug effects , Peptide Fragments/pharmacology , Plasminogen Activator Inhibitor 1/metabolism , Prolactin/pharmacology , Urokinase-Type Plasminogen Activator/antagonists & inhibitors , Animals , Cattle , Humans , Plasminogen Activator Inhibitor 1/genetics , RNA, Messenger/metabolism , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism
9.
Neuroendocrinology ; 68(6): 365-73, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9873200

ABSTRACT

Increases in the level of cAMP stimulate the secretion of GnRH from GT1 GnRH neuronal cells. We hypothesized that cyclic nucleotide phosphodiesterases (PDEs), the enzymes that hydrolyze cAMP, may constitute a negative feedback signaling mechanism for GnRH regulation by decreasing the level of cAMP. GT1 cells were shown to express three PDEs by RT-PCR analysis: the cAMP-specific PDE4B and PDE4D and the calmodulin-dependent PDE1B. A splice variant of PDE4D, PDE4D3, which is activated when phosphorylated by cAMP-dependent protein kinase (PKA), was identified in GT1 cells by Western analysis. Consistent with PDEs negatively regulating GnRH secretion, treatment with the nonselective PDE inhibitor, IBMX, stimulated GnRH secretion 137% in 30-min static cultures. Furthermore, treatment with the PDE4-specific inhibitors Rolipram and RS-25344 increased GnRH secretion 48 and 125%, while treatment with the PDE1-specific inhibitor 8-MeoM-IBMX only caused a modest increase of 28%. In perifusion studies a rapid multi-fold stimulation of GnRH secretion was observed following treatment with IBMX, Rolipram or RS-25344. In conclusion, the level of PDE activity appears to be an important negative feedback signal for GnRH secretion. We hypothesize that activation of PDE4D3 by PKA may constitute a negative feedback signaling pathway which participates in the regulation of cAMP levels.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Neurons/metabolism , Phosphoric Diester Hydrolases/metabolism , Animals , Blotting, Western , Cell Line , Cells, Cultured , Cyclic AMP/metabolism , Neurons/drug effects , Neurons/enzymology , Perfusion , Phosphodiesterase Inhibitors/pharmacology , Radioimmunoassay , Rats , Reverse Transcriptase Polymerase Chain Reaction
10.
J Cell Biochem ; 67(3): 353-66, 1997 Dec 01.
Article in English | MEDLINE | ID: mdl-9361190

ABSTRACT

Proliferation of endothelial cells is regulated by angiogenic and antiangiogenic factors whose actions are mediated by complex interactions of multiple signaling pathways. Both vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) stimulate cell proliferation and activate the mitogen-activated protein kinase (MAPK) cascade in bovine brain capillary endothelial (BBE) cells. We have extended these findings to show that both mitogens activate MAPK via stimulation of Raf-3. Activation of Raf/MAPK is inhibited by increasing intracellular cAMP levels pharmacologically or via stimulation of endogenously expressed beta-adrenergic receptors. Both VEGF- and bFGF-induced Raf-1 activity are blocked in the presence of forskolin or 8-bromo-cAMP by 80%. The actions of increased cAMP appear to be mediated by cAMP-dependent protein kinase (PKA), since treatment with H-89, a the specific inhibitor of PKA, reversed the inhibitory effect of elevated cAMP levels on mitogen-induced cell proliferation and Raf/MAPK activation. Moreover, elevations in cAMP/PKA activity inhibit mitogen-induced cell proliferation. These findings demonstrate, in cultured endothelial cells, that the cAMP/PKA signaling pathway is potentially an important physiological inhibitor of mitogen activation of the MAPK cascade and cell proliferation.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/metabolism , Fibroblast Growth Factor 2/pharmacology , Lymphokines/pharmacology , Proto-Oncogene Proteins c-raf/metabolism , Sulfonamides , 1-Methyl-3-isobutylxanthine/pharmacology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Adenylyl Cyclases/metabolism , Animals , Brain/blood supply , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Capillaries , Cattle , Cell Division , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/physiology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , Enzyme Inhibitors/pharmacology , Isoproterenol/pharmacology , Isoquinolines/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Signal Transduction/physiology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
11.
J Virol ; 71(11): 8821-31, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9343242

ABSTRACT

Neuronal death and vacuolation are characteristics of the CNS degeneration found in prion diseases. Relatively few cultured cell lines have been identified that can be persistently infected with scrapie prions, and none of these cells show cytopathologic changes reminiscent of prion neuropathology. The differentiated neuronal cell line GT1, established from gonadotropin hormone releasing-hormone neurons immortalized by genetically targeted tumorigenesis in transgenic mice (P. L. Mellon, JJ. Windle, P. C. Goldsmith, C. A. Padula, J. L. Roberts, and R. I. Weiner, Neuron 5:1-10, 1990), was examined for its ability to support prion formation. We found that GT1 cells could be persistently infected with mouse RML prions and that conditioned medium from infected cells could transfer prions to uninfected cells. In many but not all experiments, a subpopulation of cells showed reduced viability, morphological signs of neurodegeneration and vacuolation, and features of apoptosis. Subclones of GT1 cells that were stably transfected with the trk4 gene encoding the high-affinity nerve growth factor (NGF) receptor (GT1-trk) could also be persistently infected. NGF increased the viability of the scrapie-infected GT1-trk cells and reduced the morphological and biochemical signs of vacuolation and apoptosis. GT1 cells represent a novel system for studying the molecular mechanisms underlying prion infectivity and subsequent neurodegenerative changes.


Subject(s)
Apoptosis , Hypothalamus/cytology , PrPSc Proteins/metabolism , Scrapie/pathology , Animals , Apoptosis/drug effects , Cell Line , Cell Survival , DNA Fragmentation , Gene Expression , Mice , Microscopy, Electron , Models, Biological , Nerve Growth Factors/pharmacology , Vacuoles/ultrastructure
13.
Trends Endocrinol Metab ; 7(2): 65-8, 1996 Mar.
Article in English | MEDLINE | ID: mdl-18406728

ABSTRACT

The development of a functional network of gonadotropin-releasing hormone (GnRH) neurons in the central nervous system requires a series of complex regulatory mechanisms, presumably mediated in part by neurotrophic factors. The difficulty in studying factors regulating the development of GnRH neurons stems from their paucity and scattered distribution in the brain; as a result, little was known about the role of neurotrophic factors in the development of the mature GnRH neuronal network. Recent utilization of immortalized GnRH neuronal cell lines (GT1) has enabled us to identify and study specific neurotrophic factors and their functions in vitro. The potent neurotrophic effect of basic fibroblast growth factor (bFGF) and the presence of a high abundance of receptors for bFGF in GT1 cells have led to the hypothesis that bFGF may be an important regulator of GnRH neuron expansion, survival, migration, and connectivity.

14.
Endocrinology ; 136(9): 3831-8, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7649090

ABSTRACT

Basic fibroblast growth factor (bFGF) plays an important role in development of the central nervous system and is neurotropic for a variety of neurons. In this study, we investigated whether bFGF is neurotropic for GT1 GnRH neuronal cell lines and if these cells express functional FGF receptors (FGFRs). The GT1 cell lines generated by genetically targeted tumorigenesis display highly differentiated properties of GnRH neurons. Addition of 2 and 10 ng/ml bFGF increased neurite outgrowth of GT1-7 cells and resulted in a significant increase of GT1 cell survival in serum-free medium. However, bFGF had no effect on [3H]thymidine incorporation at 24 or 48 h. RNase protection assays using riboprobes specific for murine FGFRs 1-3 showed that GT1 cells express FGFRs 1 and 3 but not 2. Occupancy of FGFRs with 10 ng/ml bFGF stimulated the sustained tyrosine phosphorylation of both the 42- and 44-kilodalton mitogen-activated protein kinases (MAPKs) for up to 6 h as shown by Western blot analysis. In addition, phosphorylation of the MAPKs was associated with enzyme activation as shown by an in-gel MAPK assay. GT1-1 and GT1-7 cells also express messenger RNA for bFGF, although the level of bioactive bFGF synthesized by GT1 cells appears suboptimal because GT1 cells can further respond to exogenously added bFGF. Thus, we have demonstrated that bFGF is a neurotropic factor in GT1 GnRh neuronal cell lines, raising the possibility that bFGF may play a role in the neurobiology of GnRH neurons.


Subject(s)
Fibroblast Growth Factor 2/physiology , Gonadotropin-Releasing Hormone/metabolism , Neurons/physiology , Animals , Blotting, Western , Calcium-Calmodulin-Dependent Protein Kinases/analysis , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Cell Line , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Gonadotropin-Releasing Hormone/genetics , Mice , Mitogen-Activated Protein Kinase 1 , Neurites/physiology , Neurons/metabolism , Neurons/ultrastructure , Phosphorylation , Protein-Tyrosine Kinases/analysis , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/physiology , RNA, Messenger/analysis , RNA, Messenger/chemistry , RNA, Messenger/genetics , Thymidine/metabolism , Tritium , Tyrosine
15.
Proc Natl Acad Sci U S A ; 92(14): 6374-8, 1995 Jul 03.
Article in English | MEDLINE | ID: mdl-7541539

ABSTRACT

A number of factors both stimulating and inhibiting angiogenesis have been described. In the current work, we demonstrate that the angiogenic factor vascular endothelial growth factor (VEGF) activates mitogen-activated protein kinase (MAPK) as has been previously shown for basic fibroblast growth factor. The antiagiogenic factor 16-kDa N-terminal fragment of human prolactin inhibits activation of MAPK distal to autophosphorylation of the putative VEGF receptor, Flk-1, and phospholipase C-gamma. These data show that activation and inhibition of MAPK may play a central role in the control of angiogenesis.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/enzymology , Fibroblast Growth Factor 2/pharmacology , Lymphokines/pharmacology , Peptide Fragments/pharmacology , Prolactin/pharmacology , Animals , Blotting, Western , Capillaries , Cattle , Cells, Cultured , Cerebrovascular Circulation , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Humans , Molecular Weight , Phosphoproteins/isolation & purification , Phosphoproteins/metabolism , Phosphotyrosine , Recombinant Proteins/pharmacology , Tyrosine/analogs & derivatives , Tyrosine/analysis , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
16.
Neuroendocrinology ; 61(3): 310-7, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7898636

ABSTRACT

The GT1 GnRH neuronal cell lines exhibit highly differentiated properties of GnRH neurons. We have used GT1-1 cells to study the role of the cyclic AMP/protein kinase A, cyclic GMP/protein kinase G and Ca2+/protein kinase C signaling pathways in the regulation of GnRH secretion. Superfusion of GT1-1 cells with the cyclic AMP analog 8-Br-cyclic AMP (0.5 and 2.5 mM) or the adenylate cyclase activator forskolin (1 and 10 microM) for 100 min increased the amplitude of GnRH secretion 2- to 35-fold. The cyclic GMP analog 8-Br-cyclic GMP (2.5 mM) also stimulated the amplitude of GnRH release from superfused GT1-1 cells, although to a much lesser extent (1.5- to 3-fold). The amplitude of GnRH pulses was also stimulated (5- to 50-fold) by the protein kinase C activator TPA (1 microM). Increasing intracellular Ca2+ with an ionophore (ionomycin, 1 microM) or by the Ca2+ channel activator Bay K 8644 (10 microM) also stimulated GnRH release, while secretion was markedly decreased and spontaneous pulsatility abolished by the L-type Ca2+ channel blocker methoxyverapamil (10 microM). These results demonstrate that in GT1 cells the protein kinase A, protein kinase G and protein kinase C pathways are functionally coupled to regulation of GnRH secretion. Furthermore, pulsatile GnRH secretion is coupled to the entry of extracellular Ca2+ via L-type Ca2+ channels.


Subject(s)
Cyclic AMP/metabolism , Cyclic GMP/metabolism , Gonadotropin-Releasing Hormone/metabolism , Signal Transduction/physiology , Animals , Cell Line , Mice , Protein Kinase C/drug effects , Protein Kinase C/physiology , Secretory Rate/drug effects , Signal Transduction/drug effects
17.
J Stud Alcohol ; 55(5): 517-24, 1994 Sep.
Article in English | MEDLINE | ID: mdl-7990461

ABSTRACT

The impairing effects of alcohol on divers' abilities to perform shallow-water entry dives were investigated under controlled conditions. Each of 13 male recreational divers, 21 to 35 years old, performed three shallow-entry dives during each of seven diving sessions at various blood alcohol concentrations (BACs). The mean group BACs of Diving Sessions 1-7 were 0, 17, 40, 71, 100, 123 and 97 mg/dl, respectively. Diving performances were videotaped and rated on their relative risk of injury due to contact with the pool bottom if the depth had actually been 3.5 feet. The data indicate progressive and significant impairment of diving performance at BACs of 40 mg/dl (or 0.04 g/dl, or 0.04%) and higher. Corroborating evidence of alcohol impairment was obtained from administering Field Sobriety Tests following Sessions 1, 5, 6 and 7; almost all subjects failed the tests at the higher BAC sessions. An analysis of the self-ratings of diving performances indicated that the divers were not aware of either their degraded performance or the increased risk of injury resulting from diving after drinking.


Subject(s)
Alcoholic Intoxication , Diving , Adult , Alcohol Drinking , Humans , Male , Task Performance and Analysis , Videotape Recording , Wounds and Injuries/prevention & control
18.
Neuroendocrinology ; 59(5): 420-5, 1994 May.
Article in English | MEDLINE | ID: mdl-8022517

ABSTRACT

The mouse GT1 gonadotropin-releasing hormone (GnRH) neuronal cell lines exhibit highly differentiated properties of GnRH neurons. This report investigates the direct effect of gamma-aminobutyric acid (GABA) and subtype selective GABA agonists on GnRH secretion by GT1 cells in perifusion. Treatment of GT1-1 cells with GABA (10 microM) for 100 min resulted in a biphasic release of GnRH. A rapid and sharp stimulation of GnRH secretion was followed by a sustained inhibition of GnRH secretion. During the inhibitory phase, pulses of GnRH assessed by 'cluster analysis' were totally suppressed. The GABAA receptor agonist muscimol (10 microM) stimulated a rapid but transient release of GnRH. On the other hand, treatment of GT1-1 cells with the GABAB receptor agonist baclofen (10 microM) resulted in the prolonged inhibition of GnRH secretion which returned to normal after the treatment stopped. These results demonstrate a direct biphasic effect of GABA upon GnRH release. The initial stimulation appears to be mediated via GABAA receptors, while the sustained inhibition of GnRH secretion appears to involve the activation of GABAB receptors.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Neurons/physiology , gamma-Aminobutyric Acid/pharmacology , Animals , Baclofen/pharmacology , Cell Line , Mice , Mice, Transgenic , Muscimol/pharmacology , Neurons/drug effects , Receptors, GABA/physiology
19.
Proc Natl Acad Sci U S A ; 91(9): 3824-8, 1994 Apr 26.
Article in English | MEDLINE | ID: mdl-8170995

ABSTRACT

The result of nerve growth factor (NGF) actions depends upon the cells in which it signals. To define how signaling is influenced by cellular context, it would be useful to examine cells committed to different fates or cells of a single type at different developmental stages. Interest in NGF actions on neurons of the central nervous system led us to examine GT1-1 cells, an immortalized hypothalamic cell line. GT1-1 cells demonstrated neuronal properties but were unresponsive to NGF and other neurotrophins. Through transfection, trkA expression conferred NGF signaling leading to enhanced neuronal differentiation, including dose-dependent induction of neurite outgrowth and a rapid transient increase in c-fos and NGFI-A mRNA. Under serum-free culture conditions, NGF also delayed cell death. These findings suggest that trkA transfection of neurons and neuronal precursors can be used to better define NGF signaling.


Subject(s)
Nerve Growth Factors/pharmacology , Neurons/cytology , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Nerve Growth Factor/physiology , Amino Acid Sequence , Animals , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Line , Cross-Linking Reagents , Gene Expression/drug effects , Hypothalamus/cytology , In Vitro Techniques , Mice , Mice, Transgenic , Molecular Sequence Data , Neurites/ultrastructure , RNA, Messenger/genetics , Receptor, trkA , Transfection
20.
Proc Natl Acad Sci U S A ; 91(4): 1244-7, 1994 Feb 15.
Article in English | MEDLINE | ID: mdl-8108395

ABSTRACT

High levels of prolactin (PRL) are associated with inhibition of luteinizing hormone secretion in several mammalian species. We asked whether this phenomenon could be explained by a direct inhibitory action of PRL on hypothalamic gonadotropin hormone-releasing hormone (GnRH) neurons. The ability of PRL to suppress GnRH release and expression was tested in the highly differentiated GT1 GnRH cell lines. In static culture, nanomolar concentrations of either rat or mouse PRL inhibited the release of GnRH in a dose-dependent fashion. PRL treatment for 24 hr also decreased GnRH mRNA levels determined by Northern analysis. The cells were shown to express the PRL receptor gene, and the mRNAs for both the short and long forms were present by Northern and PCR analysis, although the short form was more abundant. In Western blots with monoclonal antibody against the rat liver PRL receptor, the short 42-kDa form of the receptor was observed. These results demonstrate that PRL inhibits GnRH release and possibly gene expression in GnRH neurons. This action appears to be mediated through prolactin receptors expressed by the cells.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Neurons/metabolism , Neurosecretory Systems/metabolism , Prolactin/pharmacology , Receptors, Prolactin/metabolism , Animals , Base Sequence , Blotting, Northern , Blotting, Western , Cell Line , Dose-Response Relationship, Drug , Gene Expression Regulation , Gonadotropin-Releasing Hormone/genetics , Luteinizing Hormone/metabolism , Mice , Molecular Sequence Data , Neurons/drug effects , Neurosecretory Systems/drug effects , Polymerase Chain Reaction , RNA, Messenger/analysis , Rats
SELECTION OF CITATIONS
SEARCH DETAIL