Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
J Neuroendocrinol ; 30(2)2018 02.
Article in English | MEDLINE | ID: mdl-28675783

ABSTRACT

Pro-opiomelanocortin (POMC) neurones within the hypothalamic arcuate nucleus are vital anorexigenic neurones. Both the insulin receptor and leptin receptor are coupled to activation of phosphatidylinositide-3 kinase (PI3K) to regulate multiple functions that increase POMC neuronal excitability. Using whole-cell recording in several mammalian species, we have found that both insulin and leptin depolarised POMC neurones via activation of transient receptor potential (TRPC)5 channels. TRPC5 channels have been rigorously characterised as the downstream effector based on their biophysical properties, pharmacological profile, and localisation by immunocytochemistry and single-cell reverse transcriptase-polymerase chain reaction. By contrast, insulin and leptin hyperpolarise and inhibit neuropeptide Y/agouti-related peptide neurones via activation of KATP channels. As proof of principle, insulin given i.c.v. robustly inhibits food intake and increases O2 utilisation, CO2 production and metabolic heat production. Therefore, these findings indicate that the depolarisation/excitation of POMC neurones by insulin and leptin is preserved across mammalian species and the activation of TRPC5 channels is likely a major mechanism by which insulin and leptin regulate energy homeostasis in mammals.


Subject(s)
Calcium Channels/metabolism , Hypothalamus/drug effects , Insulin/pharmacology , Leptin/pharmacology , Neurons/drug effects , Pro-Opiomelanocortin/metabolism , Action Potentials/drug effects , Animals , Energy Metabolism/physiology , Hypothalamus/metabolism , Neurons/metabolism
2.
J Neuroendocrinol ; 26(5): 329-40, 2014 May.
Article in English | MEDLINE | ID: mdl-24617903

ABSTRACT

Orphanin FQ/nociceptin (OFQ/N) inhibits the activity of pro-opiomelanocortin (POMC) neurones located in the hypothalamic arcuate nucleus (ARH) that regulate female sexual behaviour and energy balance. We tested the hypothesis that gonadal steroids differentially modulate the ability of OFQ/N to inhibit these cells via presynaptic inhibition of transmitter release and postsynaptic activation of G protein-gated, inwardly-rectifying K(+) (GIRK)-1 channels. Whole-cell patch clamp recordings were performed in hypothalamic slices prepared from ovariectomised rats. OFQ/N (1 µm) decreased the frequency of miniature excitatory postsynaptic currents (mEPSCs) and miniature inhibitory postsynaptic currents (mIPSCs), and also caused a robust outward current in the presence of tetrodotoxin, in ARH neurones from vehicle-treated animals. A priming dose of oestradiol benzoate (EB; 2 µg) increased basal mEPSC frequency, markedly diminished both the OFQ/N-induced decrease in mEPSC frequency and the activation of GIRK-1 currents, and potentiated the OFQ/N-induced decrease in mIPSC frequency. Steroid treatment regimens that facilitate sexual receptivity reinstate the basal mEPSC frequency, the OFQ/N-induced decrease in mEPSC frequency and the activation of GIRK-1 currents to levels observed in vehicle-treated controls, and largely abolish the ability of OFQ/N to decrease mIPSC frequency. These effects were observed in an appreciable population of identified POMC neurones, almost one-half of which projected to the medial preoptic nucleus. Taken together, these data reveal that gonadal steroids influence the pleiotropic actions of OFQ/N on ARH neurones, including POMC neurones, in a disparate manner. These temporal changes in OFQ/N responsiveness further implicate this neuropeptide system as a critical mediator of the gonadal steroid regulation of reproductive behaviour.


Subject(s)
Gonadal Steroid Hormones/pharmacology , Opioid Peptides/drug effects , Sexual Behavior, Animal/drug effects , Animals , Estradiol/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Female , Ovariectomy , Patch-Clamp Techniques , Potassium Channels/drug effects , Pro-Opiomelanocortin/physiology , Progesterone/pharmacology , Rats , Rats, Long-Evans , Receptors, Opioid/biosynthesis , Receptors, Opioid/genetics , Nociceptin Receptor , Nociceptin
3.
Am J Physiol Endocrinol Metab ; 305(5): E632-40, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23820624

ABSTRACT

Besides its quintessential role in reproduction, 17ß-estradiol (E2) is a potent anorexigenic hormone. E2 and the selective Gq-coupled membrane estrogen receptor (Gq-mER) ligand STX rapidly increase membrane excitability in proopiomelanocortin (POMC) neurons by desensitizing the coupling of GABAB receptors to G protein-coupled inwardly rectifying K(+) channels (GIRKs), which upon activation elicit a hyperpolarizing outward current. However, it is unknown whether E2 and STX can modulate GABAB signaling in neuropeptide Y (NPY)/agouti-related peptide (AgRP) neurons. We used single-cell RT-PCR and whole cell patch clamping with selective pharmacological reagents to show that NPY/AgRP cells of mice express the GABAB-R1 and -R2 receptors and are hyperpolarized by the GABAB agonist baclofen in an E2-dependent manner. In males, E2 rapidly attenuated the coupling of GABAB receptors to GIRKs, which was blocked by the general PI3K inhibitors wortmannin and LY-294002 or the selective p110ß subunit inhibitor TGX-221. The ERα-selective agonist propyl pyrazole triol mimicked the effects of E2. STX, in contrast, enhanced the GABAB response in males, which was abrogated by the estrogen receptor (ER) antagonist ICI 182,780. In gonadectomized mice of both sexes, E2 enhanced or attenuated the GABAB response in different NPY/AgRP cells. Coperfusing wortmannin with E2 or simply applying STX always enhanced the GABAB response. Thus, in NPY/AgRP neurons, activation of the Gq-mER by E2 or STX enhances the GABAergic postsynaptic response, whereas activation of ERα by E2 attenuates it. These findings demonstrate a clear functional dichotomy of rapid E2 membrane-initiated signaling via ERα vs. Gq-mER in a CNS neuron vital for regulating energy homeostasis.


Subject(s)
Acrylamides/pharmacology , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Hypothalamus/metabolism , Neurons/metabolism , Neuropeptide Y/metabolism , Receptors, GABA-B/metabolism , Acrylamides/metabolism , Animals , Baclofen/pharmacology , Estradiol/metabolism , Female , GABA-B Receptor Agonists/pharmacology , Guinea Pigs , Male , Mice , Mice, Transgenic , Patch-Clamp Techniques , Signal Transduction
4.
Biochem Soc Trans ; 33(Pt 3): 471-3, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15916543

ABSTRACT

Metazoan histone mRNAs are unique in that they lack a 3'-polyadenylated tail, but instead end in a conserved stem loop that is bound by SLBP (stem-loop binding protein). SLBP is required for efficient histone mRNA synthesis and translation. Removal of SLBP by RNA interference causes an increase in the number of cells in S-phase and this effect can be reversed by expressing an exogenous SLBP resistant to the small interfering RNA. Cells with decreased SLBP levels progress slowly through S-phase when released from a double-thymidine block. Thus SLBP is required for efficient DNA replication probably because a decreased ability to assemble chromatin results in a decrease in the rate of DNA replication.


Subject(s)
Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA Interference , S Phase , mRNA Cleavage and Polyadenylation Factors/genetics , mRNA Cleavage and Polyadenylation Factors/metabolism , Animals , Cell Line , Humans
6.
J Pharmacol Exp Ther ; 299(1): 21-30, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11561059

ABSTRACT

The present study sought to determine whether small-conductance, Ca2+-activated K+ currents underlie the afterhyperpolarization (AHP) in neurons of the preoptic area (POA), a brain region important in controlling reproduction. We used an ovariectomized, female guinea pig model to test two hypotheses: 1) the current associated with the AHP (I(AHP)) regulates the firing rate of POA neurons and 2) amine neurotransmitters modulate it in a gonadal steroid-sensitive manner. Intracellular recordings followed by combined histofluorescence/in situ hybridization for glutamic acid decarboxylase, 65-kDa isomer, revealed that POA neurons, including gamma-aminobutyric acid (GABA)ergic neurons, exhibited an AHP and spike frequency adaptation. The corresponding I(AHP) was sensitive to antagonism by CdCl2 (200 microM), apamin (0.3-1 microM), and dequalinium (3 microM). The beta-adrenergic receptor agonist isoproterenol inhibited the I(AHP) in a dose-dependent, timolol-sensitive fashion. In addition, the alpha1-adrenergic receptor agonist methoxamine dose dependently inhibited the I(AHP) in a prazosin-sensitive manner and increased neuronal firing rate. Twenty-four-hour pretreatment with estradiol benzoate (EB; 25 microg, s.c.) markedly potentiated the inhibitory effect of methoxamine on the I(AHP), whereas that for isoproterenol was unaffected. Similarly, bath application of 17beta-estradiol (100 nM; 15-20 min) mimicked the effect of EB on the methoxamine-induced inhibition of the I(AHP). Thus, POA GABAergic neurons express an apamin-sensitive channel that mediates, at least in part, the I(AHP), and tempers the excitability of these cells. Furthermore, these studies demonstrate that estrogen enhances the alpha1-adrenergic receptor-mediated inhibition of this current.


Subject(s)
Apamin/pharmacology , Estrogens/pharmacology , Hypothalamus/physiology , Neurons/metabolism , Norepinephrine/physiology , Potassium Channels, Calcium-Activated , Potassium Channels/drug effects , Reproduction/drug effects , Animals , Female , Fluorescent Dyes , Glutamate Decarboxylase/metabolism , Guinea Pigs , Hypothalamus/cytology , Hypothalamus/drug effects , In Vitro Techniques , Membrane Potentials/drug effects , Neurons/drug effects , Patch-Clamp Techniques , Preoptic Area/cytology , Preoptic Area/drug effects , Preoptic Area/physiology , Receptors, Adrenergic, alpha-1/drug effects , Receptors, Adrenergic, alpha-1/physiology , Receptors, Adrenergic, beta/drug effects , Receptors, Adrenergic, beta/physiology , Small-Conductance Calcium-Activated Potassium Channels , gamma-Aminobutyric Acid/physiology
8.
J Neurosci ; 21(6): 2085-93, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11245692

ABSTRACT

The principal role of estrogen is its control of the female ovulatory cycle via negative and positive feedback on gonadotropin secretion. However, a detailed, cohesive picture of how the steroid specifically regulates the excitability of hypothalamic neurons involved in the central control of gonadotropin secretion is still emerging. Here, we used an ovariectomized female guinea pig model to test the hypothesis that estrogen acts on GABAergic neurons in the preoptic area (POA) to elicit a biphasic profile of luteinizing hormone (LH) secretion. Intracellular electrophysiological recordings revealed that estradiol benzoate (EB; 25 microgram, s.c.) decreased the hyperpolarizing response of GABAergic neurons to the GABA(B) receptor agonist baclofen 24 hr after treatment. This effect of GABA(B) receptor stimulation in unidentified POA neurons was still depressed 42 hr after EB administration. By the use of a ribonuclease protection assay, however, EB reduced glutamic acid decarboxylase mRNA expression 42 hr but not 24 hr after its administration. Thus, estrogen attenuated the autoinhibition of GABAergic POA neurons during the initial LH suppressive (i.e., negative feedback) phase and subsequently reduced GABAergic function during the LH surge (i.e., positive feedback). These studies demonstrate that the effects of estrogen on hypothalamic GABAergic neurons coincide with the inhibitory and stimulatory actions, respectively, of the steroid on LH secretion. Furthermore, the data provide novel insights into the mechanism by which estrogen regulates hypothalamic GABAergic neurons, which are critical for the biphasic modulation of LH release observed over the course of the female ovulatory cycle.


Subject(s)
Estrogens/pharmacology , Hypothalamus/drug effects , Hypothalamus/metabolism , Luteinizing Hormone/metabolism , gamma-Aminobutyric Acid/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Dose-Response Relationship, Drug , Estradiol/analogs & derivatives , Estradiol/pharmacology , Feedback , Female , GABA Agonists/pharmacology , GABA Antagonists/pharmacology , GABA-B Receptor Agonists , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Guinea Pigs , Immunohistochemistry , In Vitro Techniques , Isoenzymes/genetics , Isoenzymes/metabolism , Luteinizing Hormone/blood , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Ovariectomy , Potassium Channels/chemistry , Potassium Channels/metabolism , Preoptic Area/cytology , Preoptic Area/drug effects , Preoptic Area/metabolism , RNA, Messenger/metabolism , Tetrodotoxin/pharmacology
9.
Mol Cell Biol ; 20(19): 7388-400, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10982855

ABSTRACT

Alternative splicing of fibroblast growth factor receptor 2 (FGF-R2) transcripts involves the mutually exclusive usage of exons IIIb and IIIc to produce two different receptor isoforms. Appropriate splicing of exon IIIb in rat prostate cancer DT3 cells requires a previously described cis element (ISAR, for "intronic splicing activator and repressor") which represses the splicing of exon IIIc and activates the splicing of exon IIIb. This element is nonfunctional in rat prostate AT3 cells, which repress exon IIIb inclusion and splice to exon IIIc. We have now identified an intronic element upstream of exon IIIb that causes repression of exon IIIb splicing. Deletion of this element abrogates the requirement for ISAR in order for exon IIIb to be spliced in DT3 cells and causes inappropriate inclusion of exon IIIb in AT3 cells. This element consists of two intronic splicing silencer (ISS) sequences, ISS1 and ISS2. The ISS1 sequence is pyrimidine rich, and in vitro cross-linking studies demonstrate binding of polypyrimidine tract binding protein (PTB) to this element. Competition studies demonstrate that mutations within ISS1 that abolish PTB binding in vitro alleviate splicing repression in vivo. Cotransfection of a PTB-1 expression vector with a minigene containing exon IIIb and the intronic splicing silencer element demonstrate PTB-mediated repression of exon IIIb splicing. Furthermore, all described PTB isoforms were equally capable of mediating this effect. Our results support a model of splicing regulation in which exon IIIc splicing does not represent a default splicing pathway but rather one in which active repression of exon IIIb splicing occurs in both cells and in which DT3 cells are able to overcome this repression in order to splice exon IIIb.


Subject(s)
Alternative Splicing/genetics , DNA-Binding Proteins/physiology , Exons/genetics , Gene Silencing , Introns/genetics , RNA-Binding Proteins/physiology , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Fibroblast Growth Factor/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Gene Expression Regulation, Neoplastic , Male , Mutagenesis, Site-Directed , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Polymerase Chain Reaction , Polypyrimidine Tract-Binding Protein , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA Precursors/metabolism , RNA, Neoplasm/metabolism , Rats , Receptor, Fibroblast Growth Factor, Type 2 , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology , Regulatory Sequences, Nucleic Acid , Sequence Deletion , Tumor Cells, Cultured
10.
Brain Res ; 879(1-2): 29-41, 2000 Oct 06.
Article in English | MEDLINE | ID: mdl-11011003

ABSTRACT

Whole-cell and intracellular recordings were made in coronal hypothalamic slices prepared from ovariectomized female guinea pigs. 62% of preoptic area (POA) neurons fired action potentials in a bursting manner, and exhibited a significantly greater afterhyperpolarization (AHP) than did non-bursting POA neurons. The majority (70%) of POA neurons (n=76) displayed a time-dependent inward rectification (I(h)) that was blocked by CsCl (3 mM) or by ZD 7288 (30 microM). In addition, 51% of the cells expressed a low-threshold spike (LTS) associated with a transient inward current (I(T)) that was blocked by NiCl(2) (200 microM). A smaller percentage of POA neurons (29%) expressed a transient outward, A-type K(+) current that was antagonized by a high concentration of 4-aminopyridine (3 mM). Moreover, POA neurons responded to bath application of the mu-opioid receptor agonist DAMGO (93%) or the GABA(B) receptor agonist baclofen (83%) with a membrane hyperpolarization or an outward current. These responses were accompanied by a decrease in input resistance or an increase in conductance, respectively, and were attenuated by BaCl(2) (100 microM). In addition, the reversal potential for these responses closely approximated the Nernst equilibrium potential for K(+). These results suggest that POA neurons endogenously express to varying degrees an AHP, an I(h), an I(T) and an A-type K(+) current. The vast majority of these neurons also are inhibited upon mu-opioid or GABA(B) receptor stimulation via the activation of an inwardly-rectifying K(+) conductance. Such intrinsic and transmitter-activated conductances likely serve as important determinants of the firing patterns of POA neurons.


Subject(s)
Action Potentials/drug effects , Neurons/physiology , Preoptic Area/physiology , 4-Aminopyridine/pharmacology , Action Potentials/physiology , Animals , Baclofen/pharmacology , Barium Compounds/pharmacology , Cesium/pharmacology , Chlorides/pharmacology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Female , Guinea Pigs , In Vitro Techniques , Membrane Potentials , Neurons/drug effects , Nickel/pharmacology , Ovariectomy , Patch-Clamp Techniques , Potassium Channels/drug effects , Potassium Channels/physiology , Preoptic Area/drug effects , Pyrimidines/pharmacology , Reaction Time , Tetrodotoxin/pharmacology
11.
Eur J Pharm Biopharm ; 50(2): 205-11, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10962228

ABSTRACT

The dose-dependent disposition, first pass hepatic elimination, and absorption pharmacokinetics (PK) of salmon calcitonin (sCT) were investigated in a canine Intestinal Vascular Access Port (IVAP) model. The PK of sCT were determined after intravenous (IV), subcutaneous (SC), portal venous (PV), and oral (PO) administration of sCT. Regional oral absorption of unformulated sCT was also evaluated by direct administration into the duodenum (ID), ileum (IL), and colon (IC) by means of surgically implanted, chronic catheters. Plasma samples were collected and analyzed by radioimmunoassay (RIA). Salmon calcitonin PK were evaluated using 2-compartmental and model independent methods. Intravenous sCT PK were non-linear over the dose range studied. High dose groups (100-1000 microg) demonstrated higher total plasma clearance (CL) and V(dss) than the low dose groups (1-25 microg). However, the MRT did not change for doses ranging from 10 to 1000 microg. After SC administration, the absorption of sCT was rapid with bioavailability (BA) varying from 21.4 to 52.9%. However, the BA of sCT was low after ID, IL, and IC administration (0.039, 0.064, and 0.021%, respectively). The role of hepatic first-pass elimination was negligible. The results of these studies demonstrate that the elimination of sCT is rapid but does not occur in the liver. Enhanced sCT clearance at higher doses was indicated by increasing V(dss) values, and it is hypothesized that increased renal blood flow and/or saturated plasma protein binding may contribute to the non-linear behavior. The IVAP canine model was found to have utility for probing the absorption and disposition PK of sCT. The combination of high oral bioavailability variability and non-linear disposition of sCT may produce highly variable therapeutic effects. The practical impact of the non-linear disposition of sCT remains to be determined. Based on the current results it appears that the rate-limiting step to the successful oral administration of sCT is its delivery into the portal vein since hepatic metabolism was negligible.


Subject(s)
Calcitonin/pharmacokinetics , Intestinal Absorption , Liver/metabolism , Administration, Oral , Animals , Dogs , Dose-Response Relationship, Drug , Male , Skin Absorption
12.
Neuroreport ; 10(12): 2681-7, 1999 Aug 20.
Article in English | MEDLINE | ID: mdl-10574392

ABSTRACT

We combined histofluorescence with in situ hybridization to identify GABAergic neurons in the arcuate nucleus (ARC) following electrophysiological recording, using GAD65 as a marker. Intracellular recordings 91 were made in hypothalamic slices prepared from ovariectomized guinea pigs. Over 90% of ARC neurons tested with the GABA(B) receptor agonist baclofen responded with a membrane hyperpolarization or an outward current. The hyperpolarization was dose dependent, and the GABA(B) receptor antagonist CGP 35,348 produced a rightward shift in the agonist dose-response curve. Agonist potency was lower, and the efficacy greater, in GAD-positive neurons. The use of this novel technique for identifying GABAergic neurons thus reveals differences in the pharmacodynamics of GABA(B) receptor activation between GABAergic and non-GABAergic ARC neurons.


Subject(s)
Arcuate Nucleus of Hypothalamus/physiology , Neural Inhibition , Neurons/physiology , Receptors, GABA-B/physiology , gamma-Aminobutyric Acid/physiology , Animals , Arcuate Nucleus of Hypothalamus/cytology , Baclofen/pharmacology , Female , Fluorescent Antibody Technique , GABA Agonists/pharmacology , GABA Antagonists/pharmacology , Glutamate Decarboxylase/analysis , Guinea Pigs , In Situ Hybridization , In Vitro Techniques , Organophosphorus Compounds/pharmacology
13.
Pharm Res ; 16(4): 527-33, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10227707

ABSTRACT

PURPOSE: To evaluate a biopharmaceutical approach for selecting formulation additives and establishing the performance specifications of an oral peptide delivery system using sCT as a model peptide. METHODS: The effect of formulation additives on sCT effective permeability and transepithelial electrical resistance (TEER) was evaluated in side-by-side diffusion chambers using rat intestinal segments. Baseline regional oral absorption of sCT was evaluated in an Intestinal and Vascular Access Port (IVAP) dog model by administration directly into the duodenum, ileum, and colon by means of surgically implanted, chronic catheters. The effect of varying the input rate and volume of the administered solution on the extent of sCT absorption was also evaluated. Citric acid (CA) was utilized in all studies to cause a transient reduction in local pH. In vitro samples and plasma samples were analyzed by radioimmunoassay (RIA). Two oral delivery systems were prepared based on the results of the in vitro and IVAP studies, and evaluated in normal dogs. RESULTS: Maximal permeability enhancement of sCT was observed using taurodeoxycholate (TDC) or lauroyl carnitine (LC) in vitro. Ileal absorption of sCT was higher than in other regions of the intestine. Low volume and bolus input of solution formulations was selected as the optimal condition for the IVAP studies since larger volumes or slower input rates resulted in significantly lower sCT bioavailability (BA). Much lower BA of sCT was observed when CA was not used in the formulation. The absolute oral bioavailability (mean+/-SD) in dogs for the control (sCT + CA) and two proprietary sCT delivery systems was 0.30%+/-0.05%, 1.10+/-0.18%, and 1.31+/-0.56%, respectively. CONCLUSIONS: These studies demonstrate the utility of in vitro evaluation and controlled in vivo studies for developing oral peptide delivery strategies. Formulation additives were selected, the optimal intestinal region for delivery identified, and the optimal release kinetics of additives and actives from the delivery system were characterized. These methods were successfully used for devising delivery strategies and fabricating and evaluating oral sCT delivery systems in animals. Based on these studies, sCT delivery systems have been fabricated and tested in humans with favorable results.


Subject(s)
Calcitonin/pharmacokinetics , Drug Delivery Systems , Intestinal Absorption/drug effects , Peptides/administration & dosage , Peptides/pharmacokinetics , Pharmaceutic Aids/pharmacology , Administration, Oral , Animals , Calcitonin/administration & dosage , Dogs , In Vitro Techniques , Male , Permeability , Rats , Rats, Sprague-Dawley , Salmon
14.
Steroids ; 64(1-2): 64-75, 1999.
Article in English | MEDLINE | ID: mdl-10323674

ABSTRACT

17Beta-estradiol (E2) rapidly (<20 min) attenuates the ability of mu-opioids to hyperpolarize guinea pig hypothalamic neurons. We have used intracellular recordings from female guinea pig hypothalamic slices to characterize the receptor and intracellular pathway(s) mediating E2's rapid effects. E2 acts stereospecifically with physiologically relevant concentration-dependence (EC50 = 8 nM) to cause a fourfold reduction in the potency of the mu-opioid agonist (D-Ala2-N-Me-Phe4-Gly5-ol)-enkephalin and the GABA(B) agonist baclofen to activate an inwardly rectifying K+ conductance in hypothalamic neurons. Both the nonsteroidal estrogen diethylstilbestrol and the anti-estrogen ICI 164,384 blocked E2 actions to uncouple mu-opioid receptors. Using a pharmacological Schild analysis, we found that ICI 164,384 competed for this E2 receptor with a Ke of approximately 0.3 nM. The protein synthesis inhibitor cycloheximide did not block the estrogenic uncoupling of the mu-opioid receptor from its K+ channel, implying a rapid, nongenomic mechanism of E2 action. The effects of E2 were mimicked by the bath application of the protein kinase A (PKA) activators, forskolin and Sp-cAMP, and the protein kinase C (PKC) activator phorbol-12,13-dibutyrate. Furthermore, the selective PKA antagonists Rp-cAMP and KT5720, which have different chemical structures and modes of action, both blocked the effects of E2. In addition, the actions of E2 were blocked by the selective PKC inhibitor Calphostin C. Therefore, it appears that E2 can activate both PKA and PKC to cause a heterologous desensitization of both mu-opioid and GABA(B) receptors, which has the potential to alter synaptic transmission in many regions of the CNS.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Estrogens/physiology , GTP-Binding Proteins/metabolism , Protein Kinase C/metabolism , Receptors, GABA-B/metabolism , Receptors, Opioid, mu/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cycloheximide/pharmacology , Enzyme Activation , Enzyme Inhibitors/pharmacology , Female , Guinea Pigs , Hypothalamus/drug effects , Hypothalamus/enzymology , Hypothalamus/metabolism , Immunohistochemistry , In Vitro Techniques , Protein Kinase C/antagonists & inhibitors , Protein Synthesis Inhibitors/pharmacology
15.
Electrophoresis ; 20(4-5): 1082-6, 1999.
Article in English | MEDLINE | ID: mdl-10344288

ABSTRACT

In this report we present evidence for a novel switch in the ratio of the two major isoforms of the polypyrimidine tract binding protein (PTB) in two related prostate cancer cell lines. The existence of different isoforms of PTB is thought to be the result of alternative splicing. We used UV cross-linking to identify a PTB doublet in the DT3 cell line, which is a rat prostate epithelial cancer line that is androgen-dependent and nonmetastatic. The AT3 cell line, a metastatic, androgen-independent cell line derived from the same tumor as the DT3 cells, was noted here to have a different isoform ratio of PTB. The two most prevalent isoforms of PTB were found to bind to an RNA probe containing a pyrimidine stretch. Western blot analysis demonstrated that these isoforms are indeed expressed differently in the two cell lines and that the observed binding is the result of this differential expression. These two cell lines are derived from the original Dunning prostate tumor, which is a model for studying tumor progression in the prostate. This ratio switch may be an important event in tumor progression in this model system of prostate cancer.


Subject(s)
Alternative Splicing , Prostatic Neoplasms/chemistry , RNA-Binding Proteins/analysis , Ribonucleoproteins/analysis , Amino Acid Sequence , Animals , Humans , Male , Molecular Sequence Data , Polypyrimidine Tract-Binding Protein , Precipitin Tests , Protein Isoforms , RNA-Binding Proteins/genetics , Rats , Ribonucleoproteins/genetics , Tumor Cells, Cultured
16.
J Pharmacol Exp Ther ; 285(3): 1266-73, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9618432

ABSTRACT

The present study examined protein kinase A (PKA) and protein kinase C (PKC) involvement in the maintenance of cellular tolerance to mu opioid receptor agonists resulting from chronic opiate exposure in neurosecretory cells of the hypothalamic arcuate nucleus (ARC). The possibility that the diminution of mu opioid receptor/effector coupling produced by acute 17beta-estradiol or chronic opiate exposures is mediated by a common kinase pathway also was investigated. Intracellular recordings were made in hypothalamic slices prepared from ovariectomized female guinea pigs. The mu opioid receptor agonist D-Ala2, N-Me-Phe4, Gly-ol5-enkephalin (DAMGO) produced dose-dependent hyperpolarizations of ARC neurons. Chronic morphine treatment for 4 days reduced DAMGO potency 2.5-fold with no change in the maximal response. This effect was mimicked by a 20-min bath application of the PKA activator cAMP, Sp-isomer, or the PKC activator phorbol-12,13-dibutyrate. A 30-min bath application of the broad-spectrum protein kinase inhibitor staurosporine completely abolished the reduced DAMGO potency seen in morphine-tolerant neurosecretory cells, including those immunopositive for gonadotropin-releasing hormone. The effect of staurosporine was mimicked by the PKA inhibitor cAMP, Rp-isomer, but not by the PKC inhibitor calphostin C. Finally, a 20-min bath application of 17beta-estradiol did not further reduce DAMGO potency in morphine-tolerant ARC neurons. Therefore, increased PKA activity maintains cellular tolerance to mu opioid receptor agonists in ARC neurosecretory cells caused by chronic morphine treatment. Furthermore, acute 17beta-estradiol and chronic opiate treatments attenuate mu opioid receptor-mediated responses via a common PKA pathway.


Subject(s)
Analgesics, Opioid/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Morphine/pharmacology , Neurosecretory Systems/drug effects , Receptors, Opioid, mu/agonists , Animals , Drug Tolerance , Enkephalin, Ala(2)-MePhe(4)-Gly(5)- , Enkephalins/pharmacology , Estrogens/metabolism , Female , Guinea Pigs , Hypothalamus/drug effects , Hypothalamus/physiology , Neurosecretory Systems/physiology , Protein Kinase C/metabolism
17.
Neuroendocrinology ; 67(2): 73-82, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9508037

ABSTRACT

Orphanin FQ (OFQ) is a novel heptadecapeptide whose structure resembles that of dynorphin A1-17. Its receptor shares appreciable homology with mu-, delta- and kappa-opioid receptors, and is highly expressed in the hypothalamus. The present study examined the effects of OFQ on neurons within the arcuate nucleus (ARC) of the mediobasal hypothalamus, using intracellular recordings from coronal slices. In current clamp, OFQ produced a hyperpolarization of ARC neurons, including those immunopositive for beta-endorphin, tyrosine hydroxylase and gonadotropin-releasing hormone. This hyperpolarization was dose-dependent, insensitive to antagonism by naloxone and was associated with a decrease in input resistance. In voltage clamp, OFQ produced an outward current associated with an increase in conductance. Varying the extracellular K+ concentration shifted the reversal potential for the OFQ response to the degree predicted by the Nernst equation. Furthermore, barium chloride markedly attenuated both the OFQ-induced hyperpolarization and decrease in input resistance. Administration of maximally effective concentrations of OFQ, followed by coadministration of maximal concentrations of either OFQ and the mu-opioid receptor agonist DAMGO or OFQ and the GABAB receptor agonist baclofen produced additive hyperpolarizations and outward currents. If DAMGO was applied first, followed by the coadministration of DAMGO and OFQ, then the responses were occluded. Taken together, these results indicate that OFQ inhibits beta-endorphin neurons, as well as A12 dopamine and GnRH neurosecretory cells, within the ARC by activating a subset of inwardly-rectifying K+ channels. This suggests that OFQ is not only an antiopioid peptide, but that it also modulates the hypothalamo-pituitary axis and, ultimately, reproductive behavior.


Subject(s)
Arcuate Nucleus of Hypothalamus/drug effects , Neurons/drug effects , Neurosecretory Systems/drug effects , Opioid Peptides/pharmacology , Potassium Channels/drug effects , beta-Endorphin/metabolism , Animals , Arcuate Nucleus of Hypothalamus/physiology , Barium Compounds/pharmacology , Chlorides/pharmacology , Electric Conductivity , Enkephalin, Ala(2)-MePhe(4)-Gly(5)- , Enkephalins/pharmacology , Female , Guinea Pigs , Neurons/physiology , Neurosecretory Systems/physiology , Potassium/pharmacology , Potassium Channel Blockers , Potassium Channels/physiology , Receptors, Opioid/agonists , Receptors, Opioid, mu/agonists , Nociceptin
18.
Neuroreport ; 9(18): 4171-7, 1998 Dec 21.
Article in English | MEDLINE | ID: mdl-9926869

ABSTRACT

We combined histofluorescence with in situ hybridization to identify GABAergic neurons in the arcuate nucleus (ARC) following electrophysiological recordings, using GAD65 as a marker. Intracellular recordings were made in hypothalamic slices prepared from ovariectomized guinea pigs. Over 90% of ARC neurons tested with the GABA(B) receptor agonist baclofen responded with a membrane hyperpolarization or an outward current. The hyperpolarization was dose-dependent, and the GABA(B) receptor antagonist CGP 35,348 produced a rightward shift in the agonist dose-response curve. Agonist potency was lower, and the efficacy greater, in GAD-positive neurons. The use of this novel technique for identifying GABAergic neurons thus reveals differences in the pharmacodynamics of GABA(B) receptor activation GABAergic and non-GABAergic ARC neurons.


Subject(s)
Arcuate Nucleus of Hypothalamus/physiology , Neural Inhibition/physiology , Neurons/physiology , Receptors, GABA-B/physiology , gamma-Aminobutyric Acid/physiology , Animals , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/drug effects , Baclofen/pharmacology , Dose-Response Relationship, Drug , Electrophysiology , Female , GABA Agonists/pharmacology , GABA Antagonists/pharmacology , Glutamate Decarboxylase/metabolism , Guinea Pigs , In Situ Hybridization, Fluorescence , In Vitro Techniques , Neurons/drug effects , Organophosphorus Compounds/pharmacology
19.
J Pharmacol Exp Ther ; 280(2): 1057-64, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9023324

ABSTRACT

The present study examined the potential for cross-tolerance development between mu-opioid and gamma-aminobutyric acidB receptor agonists, in hypothalamic arcuate neurons, resulting from chronic morphine treatment. Intracellular recordings were made in hypothalamic slices prepared from ovariectomized female guinea pigs. The mu-opioid receptor agonist D-Ala2,N-Me-Phe4,Gly-ol5-enkephalin and the gamma-aminobutyric acidB receptor agonist baclofen produced dose-dependent membrane hyperpolarizations of arcuate neurons. The reversal potential for both agonist-induced hyperpolarizations was near -95 mV, indicative of the activation of an underlying K+ conductance. Coadministration of maximally effective concentrations of D-Ala2,N-Me-Phe4,Gly-ol5-enkephalin and baclofen produced a response that was not additive, indicating a convergence onto a common K+ channel. In arcuate neurons, including a subset that was immunopositive for tyrosine hydroxylase, chronic morphine treatment for 4 to 7 days produced a 3.2-fold reduction in the potency, with no change in the efficacy, of D-Ala2,N-Me-Phe4,Gly-ol5-enkephalin. In contrast, it affected neither the potency nor the efficacy of baclofen. Therefore, chronic morphine exposure does not produce cross-tolerance between mu-opioid and gamma-aminobutyric acidB receptor agonists in A12 dopamine neurons, suggesting that convergence upon a common effector is not a sufficient criterion for the development of cross-tolerance between receptor systems.


Subject(s)
Arcuate Nucleus of Hypothalamus/physiology , Baclofen/pharmacology , Enkephalins/pharmacology , GABA-B Receptor Agonists , Hypothalamus/physiology , Morphine Dependence/physiopathology , Morphine/pharmacology , Neurons/physiology , Receptors, Opioid, mu/agonists , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Drug Tolerance , Electrophysiology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)- , Female , Guinea Pigs , Hypothalamus/drug effects , In Vitro Techniques , Membrane Potentials/drug effects , Morphine/blood , Neurons/drug effects , Tetrodotoxin/pharmacology , Tyrosine 3-Monooxygenase/analysis
20.
Neurosci Lett ; 216(2): 129-32, 1996 Sep 27.
Article in English | MEDLINE | ID: mdl-8904800

ABSTRACT

Ribonuclease protection assays were performed on the basal hypothalamus (BH), the preoptic area (POA) and the thalamus (THAL) from female guinea pigs that were ovariectomized and implanted with morphine (n = 8) or placebo (n = 8) pellets for 1 week. An antisense [32P]rUTP labeled riboprobe, representing a 280 bp fragment of the guinea pig mu-opioid receptor gene (spanning putative TM II through eight residues of TM IV), protected a single RNA band of 280 bp. In contrast to the rat, the guinea pig THAL expressed less mu-opioid receptor mRNA than both POA and BH. Morphine treatment caused a significant decrease (15.6 +/- 5.8%) in mu-opioid receptor mRNA expression in the BH, while POA and THAL were not different from placebo controls. Therefore, in conjunction with our previous findings of a downregulation of mu-opioid receptors, mu-opioid receptor mRNA is downregulated in the mediobasal hypothalamus of female guinea pigs following chronic morphine treatment.


Subject(s)
Analgesics, Opioid/pharmacology , Hypothalamus, Middle/chemistry , Morphine/pharmacology , Receptors, Opioid, mu/genetics , Analgesics, Opioid/blood , Animals , Down-Regulation/physiology , Drug Tolerance , Female , Gene Expression/drug effects , Guinea Pigs , Hypothalamus, Middle/drug effects , Hypothalamus, Middle/metabolism , Morphine/blood , Morphine Derivatives/blood , Ovariectomy , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...