Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Front Endocrinol (Lausanne) ; 12: 790441, 2021.
Article in English | MEDLINE | ID: mdl-35058881

ABSTRACT

The role of calcium, but not of other intracellular signaling molecules, in the release of pituitary hormones by exocytosis is well established. Here, we analyzed the contribution of phosphatidylinositol kinases (PIKs) to calcium-driven prolactin (PRL) release in pituitary lactotrophs: PI4Ks - which control PI4P production, PIP5Ks - which synthesize PI(4, 5)P2 by phosphorylating the D-5 position of the inositol ring of PI4P, and PI3KCs - which phosphorylate PI(4, 5)P2 to generate PI(3, 4, 5)P3. We used common and PIK-specific inhibitors to evaluate the strength of calcium-secretion coupling in rat lactotrophs. Gene expression was analyzed by single-cell RNA sequencing and qRT-PCR analysis; intracellular and released hormones were assessed by radioimmunoassay and ELISA; and single-cell calcium signaling was recorded by Fura 2 imaging. Single-cell RNA sequencing revealed the expression of Pi4ka, Pi4kb, Pi4k2a, Pi4k2b, Pip5k1a, Pip5k1c, and Pik3ca, as well as Pikfyve and Pip4k2c, in lactotrophs. Wortmannin, a PI3K and PI4K inhibitor, but not LY294002, a PI3K inhibitor, blocked spontaneous action potential driven PRL release with a half-time of ~20 min when applied in 10 µM concentration, leading to accumulation of intracellular PRL content. Wortmannin also inhibited increase in PRL release by high potassium, the calcium channel agonist Bay K8644, and calcium mobilizing thyrotropin-releasing hormone without affecting accompanying calcium signaling. GSK-A1, a specific inhibitor of PI4KA, also inhibited calcium-driven PRL secretion without affecting calcium signaling and Prl expression. In contrast, PIK93, a specific inhibitor of PI4KB, and ISA2011B and UNC3230, specific inhibitors of PIP5K1A and PIP5K1C, respectively, did not affect PRL release. These experiments revealed a key role of PI4KA in calcium-secretion coupling in pituitary lactotrophs downstream of voltage-gated and PI(4, 5)P2-dependent calcium signaling.


Subject(s)
Calcium/metabolism , Lactotrophs/metabolism , Minor Histocompatibility Antigens/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Prolactin/metabolism , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Animals , Calcium Channel Agonists/pharmacology , Calcium Signaling , Exocytosis , Lactotrophs/drug effects , Minor Histocompatibility Antigens/metabolism , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Prolactin/biosynthesis , Prolactin/drug effects , Protein Kinase Inhibitors/pharmacology , Rats , Sequence Analysis, RNA , Single-Cell Analysis , Wortmannin/pharmacology
2.
Endocrine ; 53(2): 585-94, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27038317

ABSTRACT

Prolactin is an anterior pituitary hormone necessary for fertility, pregnancy maintenance, lactation, and aspects of maternal behavior. In rodents, there is a surge of prolactin on the afternoon of proestrus, and a semi-circadian pattern of prolactin surges during early pregnancy, with a diurnal and nocturnal surge every day. Both of these patterns can be replicated in ovariectomized rats. A prior study demonstrated that central antagonism of κ-opioid receptors, the target of dynorphin, largely abolished the nocturnal prolactin surge in pregnant rats. We build on this to determine whether dynorphin, perhaps from the arcuate population that co-express kisspeptin, neurokinin B, and dynorphin (KNDy neurons), also contributes to the estradiol- or cervical stimulation-induced surges in ovariectomized rats. Ovariectomized rats were treated with either estradiol or cervical stimulation to induce prolactin surge(s). Blood samples were taken around the expected surge time to determine the effect of either acute κ-opioid receptor antagonism or previous chemical ablation of the KNDy population on prolactin levels. Dynorphin antagonism does significantly disrupt the nocturnal prolactin surge, but it does not contribute to the estradiol-induced surge. Chemical ablation of KNDy neurons had opposite effects; ablation of 40 % of the KNDy neurons had no impact on the nocturnal prolactin surge, while a somewhat larger ablation significantly reduced the size of the estradiol-induced surge. We conclude that dynorphin is likely a controlling factor for the nocturnal surge induced by cervical stimulation, and that other KNDy neuron products must play a role in the estradiol-induced surge.


Subject(s)
Dynorphins/metabolism , Estradiol/pharmacology , Estrous Cycle/drug effects , Prolactin/blood , Animals , Estrous Cycle/blood , Female , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Neurons/drug effects , Neurons/metabolism , Ovariectomy , Rats , Rats, Sprague-Dawley , Receptors, Opioid, kappa/antagonists & inhibitors
3.
Endocrinology ; 156(2): 600-12, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25406939

ABSTRACT

The peptide oxytocin (OT) is secreted by hypothalamic neurons and exerts numerous actions related to reproduction. OT stimulation of prolactin secretion in female rats is important during the estrous cycle, pregnancy, and lactation. Here we report that OT also stimulates transients of intracellular Ca(2+) concentration in somatotrophs and gonadotrophs as well as the release of GH and LH in a dose-dependent manner with EC50 values that closely correspond to the ligand affinity of the OT receptor (OTR). Remarkably, the hormone-releasing effect of OT in these two cell types is 2 orders of magnitude more sensitive than that in lactotrophs. The specific OTR agonist [Thr(4),Gly(7)]-oxytocin acutely stimulated the release of LH, GH, and prolactin from female rat pituitary cells in primary culture and increased intracellular Ca(2+) concentration in gonadotrophs, somatotrophs, and lactotrophs. In these three cell types, the effects on hormone release and intracellular Ca(2+) of both OT and [Thr(4),Gly(7)]oxytocin were abolished by the specific OT receptor antagonist desGly-NH2-d(CH2)5[D-Tyr(2),Thr(4)]OVT but not by the highly selective vasopressin V1a receptor antagonist, d(CH2)5[Tyr(Me)(2),Dab(5)]AVP. Furthermore, 10 nM arginine vasopressin stimulated LH and GH release comparably with a dose of OT that was at least 10 times lower. Finally, the presence of the OTR-like immunoreactivity could be observed in all three cell types. Taken together, these results show that OT directly stimulates gonadotrophs, somatotrophs, and lactotrophs through OT receptors and suggest that OT signaling may serve to coordinate the release of different pituitary hormones during specific physiological conditions.


Subject(s)
Oxytocin/physiology , Pituitary Gland, Anterior/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Female , Pituitary Gland, Anterior/cytology , Rats, Sprague-Dawley , Receptors, Oxytocin/metabolism
4.
Mol Endocrinol ; 26(1): 153-64, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22135067

ABSTRACT

Pituitary cells fire action potentials independently of external stimuli, and such spontaneous electrical activity is modulated by a large variety of hypothalamic and intrapituitary agonists. Here, we focused on the potential role of hyperpolarization-activated and cyclic nucleotide-gated (HCN) channels in electrical activity of cultured rat anterior pituitary cells. Quantitative RT-PCR analysis showed higher level of expression of mRNA transcripts for HCN2 and HCN3 subunits and lower expression of HCN1 and HCN4 subunits in these cells. Western immunoblot analysis of lysates from normal and GH(3) immortalized pituitary cells showed bands with appropriate molecular weights for HCN2, HCN3, and HCN4. Electrophysiological experiments showed the presence of a slowly developing hyperpolarization-activated inward current, which was blocked by Cs(+) and ZD7288, in gonadotrophs, thyrotrophs, somatotrophs, and a fraction of lactotrophs, as well as in other unidentified pituitary cell types. Stimulation of adenylyl cyclase and addition of 8-Br-cAMP enhanced this current and depolarized the cell membrane, whereas 8-Br-cGMP did not alter the current and hyperpolarized the cell membrane. Both inhibition of basal adenylyl cyclase activity and stimulation of phospholipase C signaling pathway inhibited this current. Inhibition of HCN channels affected the frequency of firing but did not abolish spontaneous electrical activity. These experiments indicate that cAMP and cGMP have opposite effects on the excitability of endocrine pituitary cells, that basal cAMP production in cultured cells is sufficient to integrate the majority of HCN channels in electrical activity, and that depletion of phosphatidylinositol 4,5-bisphosphate caused by activation of phospholipase C silences them.


Subject(s)
Cyclic Nucleotide-Gated Cation Channels/metabolism , Endocrine Cells/metabolism , Pituitary Gland, Anterior/metabolism , Potassium Channels/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Action Potentials/drug effects , Adenylyl Cyclases/biosynthesis , Animals , Cell Membrane/metabolism , Cells, Cultured , Cesium/pharmacology , Cyclic GMP/analogs & derivatives , Cyclic GMP/pharmacology , Cyclic Nucleotide-Gated Cation Channels/biosynthesis , Female , Gonadotrophs/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Lactotrophs/metabolism , Membrane Potentials/drug effects , Phosphatidylinositol 4,5-Diphosphate/deficiency , Pituitary Gland, Anterior/cytology , Potassium Channels/biosynthesis , Pyrimidines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Somatotrophs/metabolism , Thyrotrophs/metabolism , Type C Phospholipases/biosynthesis , Type C Phospholipases/metabolism
5.
J Neurosci ; 31(46): 16855-63, 2011 Nov 16.
Article in English | MEDLINE | ID: mdl-22090511

ABSTRACT

The electrical activity pattern of endocrine pituitary cells regulates their basal secretion level. Rat somatotrophs and lactotrophs exhibit spontaneous bursting and have high basal levels of hormone secretion, while gonadotrophs exhibit spontaneous spiking and have low basal hormone secretion. It has been proposed that the difference in electrical activity between bursting somatotrophs and spiking gonadotrophs is due to the presence of large conductance potassium (BK) channels on somatotrophs but not on gonadotrophs. This is one example where the role of an ion channel type may be clearly established. We demonstrate here that BK channels indeed promote bursting activity in pituitary cells. Blocking BK channels in bursting lacto-somatotroph GH4C1 cells changes their firing activity to spiking, while further adding an artificial BK conductance via dynamic clamp restores bursting. Importantly, this burst-promoting effect requires a relatively fast BK activation/deactivation, as predicted by computational models. We also show that adding a fast-activating BK conductance to spiking gonadotrophs converts the activity of these cells to bursting. Together, our results suggest that differences in BK channel expression may underlie the differences in electrical activity and basal hormone secretion levels among pituitary cell types and that the rapid rate of BK channel activation is key to its role in burst promotion.


Subject(s)
Action Potentials/physiology , Biophysical Phenomena/physiology , Ion Channel Gating/physiology , Large-Conductance Calcium-Activated Potassium Channels/physiology , Nonlinear Dynamics , Pituitary Gland/cytology , Action Potentials/drug effects , Animals , Biophysical Phenomena/drug effects , Biophysics , Cells, Cultured , Electric Conductivity , Female , Indoles/pharmacology , Ion Channel Gating/drug effects , Models, Biological , Patch-Clamp Techniques , Peptides/pharmacology , Potassium Channel Blockers/pharmacology , Rats , Rats, Sprague-Dawley , Time Factors
6.
Am J Physiol Regul Integr Comp Physiol ; 301(3): R676-81, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21677274

ABSTRACT

We have shown previously that an intravenous injection of oxytocin (OT) in ovariectomized (OVX) rats initiates a circadian rhythm of prolactin (PRL) secretion similar to that observed after cervical stimulation (CS). In this study, we investigated the pathway through which OT triggers the PRL rhythm. We first tested whether an intracerebroventricular injection of OT could trigger the PRL secretory rhythm. As it did not, we injected OT intravenously while an OT receptor antagonist was infused intravenously. This antagonist completely abolished the PRL surges, suggesting that a peripheral target of OT is necessary for triggering the PRL rhythm. We hypothesized that OT may induce PRL release, which would be transported into the brain and trigger the rhythm. In agreement with this, OT injection increased circulating PRL by 5 min. To test whether this acute increase in PRL release would induce the PRL rhythm, we compared the effect of intravenously administered thyrotropin-releasing hormone (TRH) and OT. Although TRH injection also increased PRL to a comparable level after 5 min, only OT-injected animals expressed the PRL secretory rhythm. Motivated by prior findings that bilateral resection of the pelvic nerve blocks CS-induced pseudopregnancy and OT-induced facilitation of lordosis, we then hypothesized that the OT signal may be transmitted through the pelvic nerve. In fact, OT injection failed to induce a PRL secretory rhythm in pelvic-neurectomized animals, suggesting that the integrity of the pelvic nerve is necessary for the systemic OT induction of the PRL secretory rhythm in OVX rats.


Subject(s)
Circadian Rhythm , Hypogastric Plexus/drug effects , Ovariectomy , Oxytocin/administration & dosage , Pituitary Gland, Anterior/drug effects , Prolactin/metabolism , Analysis of Variance , Animals , Denervation , Female , Hypogastric Plexus/surgery , Infusions, Intravenous , Injections, Intraventricular , Pituitary Gland, Anterior/innervation , Pituitary Gland, Anterior/metabolism , Rats , Rats, Sprague-Dawley , Thyrotropin-Releasing Hormone/administration & dosage , Time Factors
7.
Endocrinology ; 151(4): 1806-13, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20176719

ABSTRACT

Although removal of dopamine inhibition is established as a major factor in prolactin (PRL) release, a large body of evidence suggests that hypothalamic oxytocin (OT) may serve as a PRL-releasing hormone in the rat. PRL release is modulated by estradiol (E2), which rises between diestrus and proestrus of the estrous cycle, causing a PRL surge in the afternoon of proestrus. Given that E2 strongly modulates OT actions in both central and peripheral tissues, OT action on lactotrophs might also be modulated by the stage of the estrous cycle. To test this hypothesis, we have monitored PRL release and intracellular calcium levels ([Ca(2+)](i)) induced by OT in pituitary lactotrophs obtained from female rats in either diestrus 1 or proestrus. We found that both secretory and [Ca(2+)](i) responses to OT are significantly increased in lactotrophs obtained on proestrus. Moreover, we show that these differences are due to an increase in both the number of OT-responding lactotrophs and the magnitude of their individual [Ca(2+)](i) responses. Both secretory and [Ca(2+)](i) responses were abolished by a specific OT antagonist. Finally, dose-dependent studies show that the increased PRL-releasing effect of OT on proestrus is significant over a wide range of concentrations, particularly those observed in hypophyseal portal plasma. These results suggest that the rising E2 titers that culminate on proestrus facilitate the stimulatory action of OT on lactotrophs and support the notion that OT is a PRL-releasing hormone with an important role in the production of the proestrous surge of PRL.


Subject(s)
Estrous Cycle/metabolism , Lactotrophs/metabolism , Oxytocin/metabolism , Animals , Area Under Curve , Calcium/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Female , Lactotrophs/drug effects , Oxytocin/pharmacology , Prolactin/metabolism , Radioimmunoassay , Rats , Rats, Sprague-Dawley
8.
Horm Mol Biol Clin Investig ; 1(1): 35-42, 2010 Jan.
Article in English | MEDLINE | ID: mdl-25961970

ABSTRACT

BACKGROUND: Pituitary lactotrophs fire action potentials spontaneously and the associated voltage-gated calcium influx is sufficient to maintain high and steady prolactin release. Several intracellular proteins can mediate the action of calcium influx on prolactin secretion, including calmodulin-dependent protein kinases. Here, we studied effects of isoquinolonesulfonamides KN-62 and KN-93, calmodulin-dependent protein kinase inhibitors, and KN-92, an inactive analog, on spontaneous electrical activity, voltage-gated calcium influx, cyclic nucleotide production, and basal prolactin release. METHODS: The effects of these compounds on electrical activity and calcium signaling was measured in single lactotrophs and cyclic nucleotide production and prolactin release were determined in static culture and perifusion experiments of anterior pituitary cells from postpubertal female rats. RESULTS: KN-62 and KN-93 blocked basal prolactin release in a dose- and time-dependent manner, suggesting that calmodulin-dependent protein kinase could mediate the coupling of electrical activity and secretion. However, a similar effect on basal prolactin release was observed on application of KN-92, which does not inhibit this kinase. KN-93 also inhibited cAMP and cGMP production, but inhibition of prolactin release was independent of the status of cyclic nucleotide production. Single cell measurements revealed abolition of spontaneous and depolarization-induced electrical activity and calcium transients in KN-92/93-treated cells, with a time course comparable to that observed in secretory studies. CONCLUSIONS: The results suggest that caution should be used when interpreting data from studies using isoquinolonesulfonamides to evaluate the role of calmodulin-dependent protein kinases in excitable endocrine cells, because inactive compounds exhibit comparable effects on action potential secretion coupling to those of active compounds.

9.
Am J Physiol Endocrinol Metab ; 298(3): E644-51, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20009029

ABSTRACT

Anterior pituitary cells express cation-conducting P2X receptor channels (P2XRs), but their molecular identity, electrophysiological properties, cell-specific expression pattern, and physiological roles have been only partially characterized. In this study, we show by quantitative RT-PCR that mRNA transcripts for the P2X(4) subunit are the most abundant in rat anterior pituitary tissue and confirm the P2X(4)R protein expression by Western blot analysis. Single-cell patch-clamp recordings show that extracellular ATP induced an inward depolarizing current in a majority of thyrotropin-releasing hormone-responsive pituitary cells, which resembled the current profile generated by recombinant P2X(4)R. The channels were activated and desensitized in a dose-dependent manner and deactivated rapidly. Activation of these channels led to stimulation of electrical activity and promotion of voltage-gated and voltage-insensitive Ca(2+) influx. In the presence of ivermectin, a specific allosteric modulator of P2X(4)Rs, there was an approximately fourfold increase in the maximum amplitude of the ATP-induced inward current, accompanied by an increase in the sensitivity of receptors for ATP, slowed deactivation of receptors, and enhanced ATP-induced prolactin release. These results indicate that thyrotropin-releasing hormone-responsive cells, including lactotrophs, express homomeric and/or heteromeric P2X(4)Rs, which facilitate Ca(2+) influx and hormone secretion.


Subject(s)
Calcium/metabolism , Ion Channel Gating/physiology , Membrane Potentials/physiology , Pituitary Gland/cytology , Pituitary Gland/physiology , Receptors, Purinergic P2/metabolism , Animals , Cells, Cultured , Female , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2X4
10.
Ann N Y Acad Sci ; 1152: 174-86, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19161388

ABSTRACT

G(i/o) protein-coupled receptors, signaling through G protein-dependent and protein-independent pathways, have prominent effects on secretion by modulating calcium signaling and regulating the size of the releasable secretory pool, the rates of exocytosis and endocytosis, and de novo synthesis. Pituitary cells fire action potentials spontaneously, and the associated calcium influx is sufficient to maintain prolactin (PRL) release but not gonadotropin release at high and steady levels for many hours. Such secretion, termed intrinsic, spontaneous, or basal, reflects fusion of secretory vesicles triggered by the cell type-specific pattern of action potentials. In lactotrophs, activation of endothelin ET(A) and dopamine D(2) receptors causes inhibition of spontaneous electrical activity and basal adenylyl cyclase activity accompanied with inhibition of basal PRL release. Agonist-induced inhibition of cAMP production and firing of action potentials is abolished in cells with blocked pertussis toxin (PTX)-sensitive G(i/o) signaling pathway. However, agonist-induced inhibition of PRL release is only partially relieved in such treated cells, indicating that both receptors also inhibit exocytosis downstream of cAMP/calcium signaling. The PTX-insensitive step in agonist-induced inhibition of PRL release is not affected by inhibition of phosphoinositide 3-kinase and glycogen synthase kinase-3 but is partially rescued by downregulation of the G(z)alpha expression. Thus, ET(A) and D(2) receptors inhibit basal PRL release not only by blocking electrical activity but also by desensitizing calcium-secretion coupling.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Lactotrophs/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Humans , Prolactin/metabolism , Protein Binding , Signal Transduction
11.
Endocrinology ; 149(4): 1470-9, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18096663

ABSTRACT

Dopamine D2 receptors signal through the pertussis toxin (PTX)-sensitive G(i/o) and PTX-insensitive G(z) proteins, as well as through a G protein-independent, beta-arrestin/glycogen synthase kinase-3-dependent pathway. Activation of these receptors in pituitary lactotrophs leads to inhibition of prolactin (PRL) release. It has been suggested that this inhibition occurs through the G(i/o)-alpha protein-mediated inhibition of cAMP production and/or G(i/o)-betagamma dimer-mediated activation of inward rectifier K(+) channels and inhibition of voltage-gated Ca(2+) channels. Here we show that the dopamine agonist-induced inhibition of spontaneous Ca(2+) influx and release of prestored PRL was preserved when cAMP levels were elevated by forskolin treatment. We further observed that dopamine agonists inhibited both spontaneous and depolarization-induced Ca(2+) influx in untreated but not in PTX-treated cells. This inhibition was also observed in cells with blocked inward rectifier K(+) channels, suggesting that the dopamine effect on voltage-gated Ca(2+) channel gating is sufficient to inhibit spontaneous Ca(2+) influx. However, agonist-induced inhibition of PRL release was only partially relieved in PTX-treated cells, indicating that dopamine receptors also inhibit exocytosis downstream of voltage-gated Ca(2+) influx. The PTX-insensitive step in agonist-induced inhibition of PRL release was not affected by the addition of wortmannin, an inhibitor of phosphatidylinositol 3-kinase, and lithium, an inhibitor of glycogen synthase kinase-3, but was attenuated in the presence of phorbol 12-myristate 13-acetate, which inhibits G(z) signaling pathway in a protein kinase C-dependent manner. Thus, dopamine inhibits basal PRL release by blocking voltage-gated Ca(2+) influx through the PTX-sensitive signaling pathway and by desensitizing Ca(2+) secretion coupling through the PTX-insensitive and protein kinase C-sensitive signaling pathway.


Subject(s)
Dopamine/pharmacology , Pertussis Toxin/pharmacology , Pituitary Gland/metabolism , Prolactin/metabolism , Signal Transduction/physiology , Adenylyl Cyclase Inhibitors , Androstadienes/pharmacology , Animals , Calcium/metabolism , Cells, Cultured , Cyclic AMP/biosynthesis , Female , Lithium/pharmacology , Potassium Channels, Inwardly Rectifying/physiology , Rats , Rats, Sprague-Dawley , Tetradecanoylphorbol Acetate/pharmacology , Wortmannin
12.
Biochem Biophys Res Commun ; 346(3): 845-50, 2006 Aug 04.
Article in English | MEDLINE | ID: mdl-16780797

ABSTRACT

Pituitary lactotrophs fire action potentials spontaneously and the associated voltage-gated calcium influx is sufficient to maintain high prolactin release. Here we studied the role of hyperpolarization-activated cation channels in pacemaking activity, calcium signaling, and prolactin secretion in these cells. A slowly developing and hyperpolarization-activated inward current was identified but only in a fraction of lactotrophs. The current was blocked by ZD7288, a relatively specific blocker of these channels. However, the pacemaking activity increased in ZD7288-treated cells independently of the presence of this current. This in turn facilitated voltage-gated calcium influx and transiently stimulated prolactin secretion. Sustained ZD7288 application in concentrations that are commonly used to block the hyperpolarization-activated cation channels inhibited hormone release at elevated intracellular calcium concentrations. Agonist and Bay K 8644-stimulated prolactin release was also inhibited by ZD7288, indicating that this compound attenuates the exocytotic pathway downstream of calcium influx.


Subject(s)
Calcium/metabolism , Exocytosis/drug effects , Ion Channels/metabolism , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Pyrimidines/pharmacology , Animals , Cations, Divalent/metabolism , Cells, Cultured , Cyclic Nucleotide-Gated Cation Channels , Electrophysiology , Female , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Patch-Clamp Techniques , Pituitary Gland/cytology , Potassium Channels , Rats , Rats, Sprague-Dawley
13.
Mol Endocrinol ; 20(9): 2231-46, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16645040

ABSTRACT

Pituitary lactotrophs in vitro fire extracellular Ca2+-dependent action potentials spontaneously through still unidentified pacemaking channels, and the associated voltage-gated Ca2+influx (VGCI) is sufficient to maintain basal prolactin (PRL) secretion high and steady. Numerous plasma membrane channels have been characterized in these cells, but the mechanism underlying their pacemaking activity is still not known. Here we studied the relevance of cyclic nucleotide signaling pathways in control of pacemaking, VGCI, and PRL release. In mixed anterior pituitary cells, both VGCI-inhibitable and -insensitive adenylyl cyclase (AC) subtypes contributed to the basal cAMP production, and soluble guanylyl cyclase was exclusively responsible for basal cGMP production. Inhibition of basal AC activity, but not soluble guanylyl cyclase activity, reduced PRL release. In contrast, forskolin stimulated cAMP and cGMP production as well as pacemaking, VGCI, and PRL secretion. Elevation in cAMP and cGMP levels by inhibition of phosphodiesterase activity was also accompanied with increased PRL release. The AC inhibitors attenuated forskolin-stimulated cyclic nucleotide production, VGCI, and PRL release. The cell-permeable 8-bromo-cAMP stimulated firing of action potentials and PRL release and rescued hormone secretion in cells with inhibited ACs in an extracellular Ca2+-dependent manner, whereas 8-bromo-cGMP and 8-(4-chlorophenylthio)-2'-O-methyl-cAMP were ineffective. Protein kinase A inhibitors did not stop spontaneous and forskolin-stimulated pacemaking, VGCI, and PRL release. These results indicate that cAMP facilitates pacemaking, VGCI, and PRL release in lactotrophs predominantly in a protein kinase A- and Epac cAMP receptor-independent manner.


Subject(s)
Adenylyl Cyclases/metabolism , Calcium/metabolism , Pituitary Gland/metabolism , Prolactin/metabolism , Signal Transduction , Adenylyl Cyclase Inhibitors , Animals , Calcium Channels/metabolism , Cell Membrane Permeability/drug effects , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Electron Transport , Electrophysiology , Enzyme Inhibitors/pharmacology , Female , Ion Channel Gating , Patch-Clamp Techniques , Phosphoric Diester Hydrolases/metabolism , Pituitary Gland/cytology , Pituitary Gland/drug effects , Rats , Rats, Sprague-Dawley
14.
J Biol Chem ; 280(29): 26896-903, 2005 Jul 22.
Article in English | MEDLINE | ID: mdl-15919662

ABSTRACT

The G protein-coupled receptors in excitable cells have prominent roles in controlling Ca2+-triggered secretion by modulating voltage-gated Ca2+ influx. In pituitary lactotrophs, spontaneous voltage-gated Ca2+ influx is sufficient to maintain prolactin release high. Here we show that endothelin in picomolar concentrations can interrupt such release for several hours downstream of spontaneous and high K+-stimulated voltage-gated Ca2+ influx. This action occurred through the Gz signaling pathway; the adenylyl cyclase-signaling cascade could mediate sustained inhibition of secretion, whereas rapid inhibition also occurred at elevated cAMP levels regardless of the status of phospholipase C, tyrosine kinases, and protein kinase C. In a nanomolar concentration range, endothelin also inhibited voltage-gated Ca2+ influx through the G i/o signaling pathway. Thus, the coupling of seven-transmembrane domain endothelin receptors to Gz proteins provided a pathway that effectively blocked hormone secretion distal to Ca2+ entry, whereas the cross-coupling to G i/o proteins reinforced such inhibition by simultaneously reducing the pacemaking activity.


Subject(s)
Calcium/metabolism , Endothelin-1/pharmacology , GTP-Binding Protein alpha Subunits/metabolism , Pituitary Gland, Anterior/metabolism , Adenylyl Cyclases/metabolism , Animals , Calcium Channels/drug effects , Female , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Pituitary Gland, Anterior/cytology , Prolactin/metabolism , Protein Subunits , Rats , Rats, Sprague-Dawley , Receptors, Endothelin/metabolism , Receptors, G-Protein-Coupled/physiology , Signal Transduction
15.
Purinergic Signal ; 1(2): 135-44, 2005 Jun.
Article in English | MEDLINE | ID: mdl-18404498

ABSTRACT

Hypothalamic and pituitary cells express G protein-coupled adenosine and P2Y receptors and cation-conducting P2X receptor-channels, suggesting that extracellular ATP and other nucleotides may function as autocrine and/or paracrine signaling factors in these cells. Consistent with this hypothesis, we show that cultured normal and immortalized pituitary and hypothalamic cells release ATP under resting conditions. RT-PCR analysis also revealed the presence of transcripts for ecto-nucleotidase eNTPDase 1-3 in these cells. These enzymes were functional as documented by degradation of endogenously released and exogenously added ATP. Blocking the activity of eNTPDases by ARL67156 led to an increase in ATP release in perifused pituitary cells and inhibition of degradation of extracellularly added ATP. Furthermore, the addition of apyrase, a soluble ecto-nucleotidase, and the expression of recombinant mouse eNTPDase-2, enhanced degradation of both endogenously released and exogenously added ATP. The released ATP by resting hypothalamic cells was sufficient to activate and desensitize high-affinity recombinant P2X receptors, whereas facilitation of ATP metabolism by the addition of apyrase protected their desensitization. These results indicate that colocalization of ATP release sites and ecto-nucleotidase activity at the plasma membrane of hypothalamic and pituitary cells provides an effective mechanism for the operation of nucleotides as extracellular signaling molecules.

16.
J Biol Chem ; 278(47): 46270-7, 2003 Nov 21.
Article in English | MEDLINE | ID: mdl-12970352

ABSTRACT

Anterior pituitary cells express nucleotide-gated G protein-coupled P2 receptors (P2YRs) and cation-conducting channels (P2XRs). However, the identification of P2 receptors subtypes and their native ligands, and the distribution and function of these receptors within the secretory and non-secretory pituitary cells has been incompletely characterized. The focus in this study was on lactotroph subpopulation of cells. ATP and ADP, but not UTP and UDP, triggered calcium signaling in a majority (85%) of lactotrophs and prolactin release in mixed pituitary cells. Consistent with the role of P2 receptors in signaling and secretion, the actions of ATP and ADP were abolished in the presence of apyrase, an ectonucleotidase. Transcripts for Gq-coupled calcium-mobilizing P2Y1R, P2Y2R, P2Y4R, and P2Y6R, as well as Gi-coupled P2Y12R, were identified in mixed anterior pituitary cells. The ligand-selectivity profile of calcium mobilization-dependent signaling and prolactin secretion and the blockade of these responses by pyridoxal 5-phosphate 6-azophenyl-2',4'-disulphonic acid indicated that P2Y1R mediates the stimulatory action of ATP and ADP. Within the channels expressed in anterior pituitary (P2X2R, P2X3R, P2X4R, and P2X7R), the P2X4R subtype provides a major pathway for calcium influx-dependent signaling and prolactin secretion. This conclusion was based on comparison of native to recombinant channels with respect to their ligand preference, sensitivity to pyridoxal 5-phosphate 6-azophenyl-2',4'-disulphonic acid, and the rates of calcium signal desensitization.


Subject(s)
Calcium Signaling , Pituitary Gland, Anterior/cytology , Prolactin/metabolism , Receptors, Purinergic P2/physiology , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Calcium Signaling/drug effects , Cell Line , Female , Ligands , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X4 , Receptors, Purinergic P2Y1
17.
Endocrinology ; 144(7): 2912-21, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12810546

ABSTRACT

The coupling between nitric oxide (NO)-cGMP signaling pathway and prolactin (PRL) release in pituitary lactotrophs has been established previously. However, the messenger that mediates the action of this signaling pathway on hormone secretion and the secretory mechanism affected, calcium dependent or independent, have not been identified. In cultured pituitary cells, basal PRL release was controlled by spontaneous voltage-gated calcium influx and was further enhanced by depolarization of cells and stimulation with TRH. Inhibition of constitutively expressed neuronal NO synthase decreased NO and cGMP levels and increased basal PRL release. The addition of a slowly releasable NO donor increased cGMP levels and inhibited basal PRL release in a time-dependent manner. Expression of inducible NO synthase also increased NO and cGMP levels and inhibited basal, depolarization-induced, and TRH-induced PRL release, whereas inhibition of this enzyme decreased NO and cGMP production and recovered PRL release. None of these treatments affected spontaneous and stimulated voltage-gated calcium influx. At basal NO levels, the addition of permeable cGMP analogs did not inhibit PRL secretion. At elevated NO levels, inhibition of cGMP production and facilitation of its degradation did not reverse inhibited PRL secretion. These experiments indicate that NO inhibits calcium-dependent PRL secretion in a cGMP-independent manner and downstream of voltage-gated calcium influx.


Subject(s)
Calcium/metabolism , Nitric Oxide/metabolism , Pituitary Gland, Anterior/metabolism , Prolactin/metabolism , Animals , Calcium Channels/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Cells, Cultured , Cyclic GMP/metabolism , Female , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type II , Patch-Clamp Techniques , Pituitary Gland, Anterior/cytology , Rats , Rats, Sprague-Dawley , Thyrotropin-Releasing Hormone/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...