Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Int J Obes (Lond) ; 39(1): 105-13, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24732144

ABSTRACT

BACKGROUND: In the central nervous system (CNS), thyrotropin-releasing hormone (TRH) has an important role in regulating energy balance. We previously showed that dietary deprivation of leucine in mice increases energy expenditure through CNS-dependent regulation. However, the involvement of central TRH in this regulation has not been reported. METHODS: Male C57J/B6 mice were maintained on a control or leucine-deficient diet for 7 days. Leucine-deprived mice were either third intracerebroventricular (i.c.v.) injected with a TRH antibody followed by intraperitoneal (i.p.) injection of triiodothyronine (T3) or i.c.v. administrated with an adenovirus of shCREB (cAMP-response element binding protein) followed by i.c.v. injection of TRH. Food intake and body weight were monitored daily. Oxygen consumption, physical activity and rectal temperature were assessed after the treatment. After being killed, the hypothalamus and the brown adipose tissue were collected and the expression of related genes and proteins related was analyzed. In other experiments, control or leucine-deficient medium incubated primary cultured neurons were either infected with adenovirus-mediated short hairpin RNA targeting extracellular signal-regulated kinases 1 and 2 (Ad-shERK1/2) or transfected with plasmid-overexpressing protein phosphatase 1 regulatory subunit 3C (PPP1R3C). RESULTS: I.c.v. administration of anti-TRH antibodies significantly reduced leucine deprivation-stimulated energy expenditure. Furthermore, the effects of i.c.v. TRH antibodies were reversed by i.p. injection of T3 during leucine deprivation. Moreover, i.c.v. injection of Ad-shCREB (adenovirus-mediated short hairpin RNA targeting CREB) significantly suppressed leucine deprivation-stimulated energy expenditure via modulation of TRH expression. Lastly, TRH expression was regulated by CREB, which was phosphorylated by ERK1/2 and dephosphorylated by PPP1R3C-containing protein Ser/Thr phosphatase type 1 (PP1) under leucine deprivation in vitro. CONCLUSIONS: Our data indicate a novel role for TRH in regulating energy expenditure via T3 during leucine deprivation. Furthermore, our findings reveal that TRH expression is activated by CREB, which is phosphorylated by ERK1/2 and dephosphorylated by PPP1R3C-containing PP1. Collectively, our studies provide novel insights into the regulation of energy homeostasis by the CNS in response to an essential amino-acid deprivation.


Subject(s)
Central Nervous System/metabolism , Energy Metabolism/drug effects , Hypothalamus/metabolism , Leucine/deficiency , Receptors, Thyrotropin-Releasing Hormone/metabolism , Thyrotropin-Releasing Hormone/metabolism , Animals , Antibodies/pharmacology , Blotting, Western , Central Nervous System/physiopathology , Hypothalamus/physiopathology , Injections, Intraventricular , Male , Mice , Mice, Inbred C57BL , Receptors, Thyrotropin-Releasing Hormone/drug effects , Signal Transduction
2.
Mol Cell Biol ; 26(6): 2247-61, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16508001

ABSTRACT

In pituitary GH3B6 cells, signaling involving the protein kinase C (PKC) multigene family can self-organize into a spatiotemporally coordinated cascade of isoform activation. Indeed, thyrotropin-releasing hormone (TRH) receptor activation sequentially activated green fluorescent protein (GFP)-tagged or endogenous PKCbeta1, PKCalpha, PKCepsilon, and PKCdelta, resulting in their accumulation at the entire plasma membrane (PKCbeta and -delta) or selectively at the cell-cell contacts (PKCalpha and -epsilon). The duration of activation ranged from 20 s for PKCalpha to 20 min for PKCepsilon. PKCalpha and -epsilon selective localization was lost in the presence of Gö6976, suggesting that accumulation at cell-cell contacts is dependent on the activity of a conventional PKC. Constitutively active, dominant-negative PKCs and small interfering RNAs showed that PKCalpha localization is controlled by PKCbeta1 activity and is calcium independent, while PKCepsilon localization is dependent on PKCalpha activity. PKCdelta was independent of the cascade linking PKCbeta1, -alpha, and -epsilon. Furthermore, PKCalpha, but not PKCepsilon, is involved in the TRH-induced beta-catenin relocation at cell-cell contacts, suggesting that PKCepsilon is not the unique functional effector of the cascade. Thus, TRH receptor activation results in PKCbeta1 activation, which in turn initiates a calcium-independent but PKCbeta1 activity-dependent sequential translocation of PKCalpha and -epsilon. These results challenge the current understanding of PKC signaling and raise the question of a functional dependence between isoforms.


Subject(s)
Protein Kinase C/metabolism , Signal Transduction , Animals , Calcium/metabolism , Carbazoles/pharmacology , Cell Communication , Cell Line , Enzyme Activation , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Ionomycin/pharmacology , Isoenzymes/genetics , Isoenzymes/metabolism , Pituitary Gland/cytology , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/drug effects , Protein Kinase C/genetics , Protein Kinase C beta , Protein Transport , Rats , Receptors, Thyrotropin-Releasing Hormone/drug effects , Receptors, Thyrotropin-Releasing Hormone/metabolism , Thyrotropin-Releasing Hormone/metabolism , Thyrotropin-Releasing Hormone/pharmacology , beta Catenin/drug effects , beta Catenin/metabolism
3.
Cell Tissue Res ; 334(2): 283-94, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18795335

ABSTRACT

Thyrotropin-releasing hormone (TRH) was initially discovered as a neuropeptide synthesized in the hypothalamus. Receptors for this hormone include TRH-receptor-1 (TRH-R1) and -2 (TRH-R2). Previous studies have shown that TRH-R1 and TRH-R2 are localized exclusively in adult Leydig cells (ALCs). We have investigated TRH-R1 and TRH-R2 expression in the testes of postnatal 8-, 14-, 21- 35-, 60-, and 90-day-old rats and in ethane dimethane sulfonate (EDS)-treated adult rats by using Western blotting, immunohistochemistry, and immunofluorescence. The effects of TRH on testosterone secretion of primary cultured ALCs from 90-day-old rats and DNA synthesis in Leydig cells from 21-day-old rats have also been examined. Western blotting and immunohistochemistry demonstrated that TRH-R1 and TRH-R2 were expressed in fetal Leydig cells (in 8-day-old rats) and in all stages of adult-type Leydig cells during development. Immunofluorescence double-staining revealed that newly regenerated Leydig cells in post-EDS 21-day rats expressed TRH-R1 and TRH-R2 on their first reappearance. Incubation with various doses of TRH affected testosterone secretion of primary cultured ALCs. Low concentrations of TRH (0.001, 0.01, and 0.1 ng/ml) inhibited basal and human chorionic gonadotrophin (hCG)-stimulated testosterone secretion of isolated ALCs, whereas relatively high doses of TRH (1 and 10 ng/ml) increased hCG-stimulated testosterone secretion. As detected by a 5-bromo-2'-deoxyuridine incorporation test, the DNA synthesis of Leydig cells from 21-day-old rats was promoted by low TRH concentrations. Thus, we have clarified the effect of TRH on testicular function: TRH might regulate the development of Leydig cells before maturation and the secretion of testosterone after maturation.


Subject(s)
Leydig Cells/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Thyrotropin-Releasing Hormone/metabolism , Testosterone/biosynthesis , Animals , Hormones/pharmacology , Leydig Cells/drug effects , Leydig Cells/ultrastructure , Male , Mesylates/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/drug effects , Receptors, Thyrotropin-Releasing Hormone/drug effects , Testis/drug effects , Testis/metabolism , Testis/ultrastructure , Thyrotropin-Releasing Hormone/pharmacology
4.
Neurosci Lett ; 431(1): 26-30, 2008 Jan 24.
Article in English | MEDLINE | ID: mdl-18069127

ABSTRACT

Glp-Asn-Pro-D-Tyr-D-TrpNH(2) is a novel synthetic peptide that mimics and amplifies central actions of thyrotropin-releasing hormone (TRH) in rat without releasing TSH. The aim of this study was to compare the binding properties of this pentapeptide and its all-L counterpart (Glp-Asn-Pro-Tyr-TrpNH(2)) to TRH receptors in native rat brain tissue and cells expressing the two TRH receptor subtypes identified in rat to date, namely TRHR1 and TRHR2. Radioligand binding studies were carried out using [(3)H][3-Me-His(2)]TRH to label receptors in hippocampal, cortical and pituitary tissue, GH4 pituitary cells, as well as CHO cells expressing TRHR1 and/or TRHR2. In situ hybridization studies suggest that cortex expresses primarily TRHR2 mRNA, hippocampus primarily TRHR1 mRNA and pituitary exclusively TRHR1 mRNA. Competition experiments showed [3-Me-His(2)]TRH potently displaced [(3)H][3-Me-His(2)]TRH binding from all tissues/cells investigated. Glp-Asn-Pro-D-Tyr-D-TrpNH(2) in concentrations up to 10(-5)M did not displace [(3)H][3-Me-His(2)]TRH binding to membranes derived from GH4 cells or CHO-TRHR1 cells, consistent with its lack of binding to pituitary membranes and TSH-releasing activity. Similar results were obtained for the corresponding all-L peptide. In contrast, both pentapeptides displaced binding from rat hippocampal membranes (pIC(50) Glp-Asn-Pro-D-Tyr-D-TrpNH(2): 7.7+/-0.2; pIC(50) Glp-Asn-Pro-Tyr-TrpNH(2): 6.6+/-0.2), analogous to cortical membranes (pIC(50) Glp-Asn-Pro-D-Tyr-D-TrpNH(2): 7.8+/-0.2; pIC(50) Glp-Asn-Pro-Tyr-TrpNH(2): 6.6+/-0.2). Neither peptide, however, displaced [(3)H][3-Me-His(2)]TRH binding to CHO-TRHR2. Thus, this study reveals for the first time significant differences in the binding properties of native and heterologously expressed TRH receptors. Also, the results raise the possibility that Glp-Asn-Pro-D-Tyr-D-TrpNH(2) is not displacing [(3)H][3-Me-His(2)]TRH from a known TRH receptor in rat cortex, but rather a hitherto unidentified TRH receptor.


Subject(s)
Cerebral Cortex/metabolism , Hippocampus/metabolism , Oligopeptides/metabolism , Pituitary Gland/metabolism , Receptors, Thyrotropin-Releasing Hormone/metabolism , Thyrotropin-Releasing Hormone/analogs & derivatives , Thyrotropin-Releasing Hormone/metabolism , Amino Acid Sequence/physiology , Animals , Binding Sites/physiology , Binding, Competitive/drug effects , Binding, Competitive/physiology , CHO Cells , Cerebral Cortex/drug effects , Cricetinae , Cricetulus , Hippocampus/drug effects , Oligopeptides/chemical synthesis , Oligopeptides/pharmacology , Peptide Fragments/chemical synthesis , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Peptide Hormones/metabolism , Peptide Hormones/pharmacology , Pituitary Gland/drug effects , Radioligand Assay , Rats , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/metabolism , Receptors, Thyrotropin-Releasing Hormone/drug effects , Thyrotropin-Releasing Hormone/chemical synthesis , Thyrotropin-Releasing Hormone/pharmacology
5.
Cell Biochem Funct ; 26(2): 264-74, 2008.
Article in English | MEDLINE | ID: mdl-18041110

ABSTRACT

The molecular mechanisms involved in GPCR-initiated signaling cascades where the two receptors share the same signaling cascade, such as thyrotropin-releasing hormone (TRH) and angiotensin II (ANG II), are still far from being understood. Here, we analyzed hormone-induced Ca(2+) responses and the process of desensitization in HEK-293 cells, which express endogenous ANG II receptors. These cells were transfected to express exogenously high levels of TRH receptors (clone E2) or both TRH receptors and G(11)alpha protein (clone E2M11). We observed that the characteristics of the Ca(2+) response, as well as the process of desensitization, were both strongly dependent on receptor number and G(11)alpha protein level. Whereas treatment of E2 cells with TRH or ANG II led to significant desensitization of the Ca(2+) response to subsequent addition of either hormone, the response was not desensitized in E2M11 cells expressing high levels of G(11)alpha. In addition, stimulation of both cell lines with THR elicited a clear heterologous desensitization to subsequent stimulation with ANG II. On the other hand, ANG II did not affect a subsequent response to TRH. ANG II-mediated signal transduction was strongly dependent on plasma membrane integrity modified by cholesterol depletion, but signaling through TRH receptors was altered only slightly under these conditions. It may be concluded that the level of expression of G-protein-coupled receptors and their cognate G-proteins strongly influences not only the magnitude of the Ca(2+) response but also the process of desensitization and resistance to subsequent hormone addition.


Subject(s)
Angiotensin II/pharmacology , Calcium Signaling/drug effects , Calcium/metabolism , Cell Membrane/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/biosynthesis , Thyrotropin-Releasing Hormone/pharmacology , Calcium/pharmacology , Calcium Signaling/physiology , Cell Membrane/drug effects , Cells, Cultured , Cholesterol/analysis , Cholesterol/metabolism , Dose-Response Relationship, Drug , GTP-Binding Protein alpha Subunits, Gq-G11/drug effects , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Humans , Receptors, Thyrotropin-Releasing Hormone/biosynthesis , Receptors, Thyrotropin-Releasing Hormone/drug effects , Temperature , Time Factors , Transfection
6.
Mol Endocrinol ; 19(11): 2859-70, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16020481

ABSTRACT

To investigate the function of dimerization of the TRH receptor, a controlled dimerization system was developed. A variant FK506 binding protein (FKBP) domain was fused to the receptor C terminus and dimerization induced by incubating cells with dimeric FKBP ligand, AP20187. The TRH receptor-fusion bound hormone and signaled normally. Addition of dimerizer to cells expressing the receptor-FKBP fusion dramatically increased the fraction of receptor running as dimer on SDS-PAGE. AP20187 caused dimerization in a time- and concentration-dependent manner, acting within 1 min. Dimerizer had no effect on TRH receptors lacking the FKBP domain, and its effects were blocked by excess monomeric FKBP ligand. AP20187-induced dimerization did not cause receptor phosphorylation, inositol phosphate production, or ERK1/2 activation, and dimerizer did not alter signaling by TRH. Induced dimerization did, however, alter TRH receptor trafficking. TRH promoted greater receptor internalization in cells treated with AP20187 but not monomeric ligand, based on loss of surface binding sites and immunostaining. Dimerization increased the rate of internalization of TRH receptors and decreased the apparent rate of receptor recycling. AP20187 enhanced the small amount of TRH-induced receptor internalization when the receptor-FKBP fusion protein was expressed in cells lacking beta-arrestins. The results show that controlled dimerization of the TRH receptor potentiates hormone-induced receptor trafficking.


Subject(s)
Receptors, Thyrotropin-Releasing Hormone/metabolism , Thyrotropin-Releasing Hormone/metabolism , Animals , Arrestins/metabolism , CHO Cells , Calcium/metabolism , Cell Membrane/chemistry , Cricetinae , Dimerization , Enzyme Activation/drug effects , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Protein Transport/drug effects , Receptors, Thyrotropin-Releasing Hormone/analysis , Receptors, Thyrotropin-Releasing Hormone/drug effects , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/metabolism , Signal Transduction/drug effects , Tacrolimus/analogs & derivatives , Tacrolimus/pharmacology , Tacrolimus Binding Proteins/analysis , Tacrolimus Binding Proteins/drug effects , Tacrolimus Binding Proteins/metabolism , Thyrotropin-Releasing Hormone/pharmacology , Type C Phospholipases/metabolism , beta-Arrestins
7.
Arch Gen Psychiatry ; 51(11): 875-83, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7944876

ABSTRACT

BACKGROUND: A reduced thyrotropin (TSH) response to thyrotropin-releasing hormone (protirelin [TRH]) has been found consistently in a portion of patients with major depression. One hypothesis to explain this observation is that pituitary TRH receptors are down-regulated in major depression. One prediction stemming from this hypothesis is that prolactin (PRL) as well as TSH responses to TRH should be attenuated. To adequately test the pattern of protirelin-induced TSH and PRL responses with a protirelin dose-response design is necessary. METHODS: Four doses of protirelin (25, 100, 500, and 800 micrograms) were infused in an ascending schedule at intervals of 3 to 7 days in patients with major depression and in control subjects. Seven women and six men with major depression were compared with age- and gender-matched controls (five women and seven men). The TSH and PRL responses were measured at regular intervals following each dose of protirelin. RESULTS: No significant group differences in baseline levels of thyroid hormones or cortisol were present. Depressed men exhibited significant reductions in both TSH and PRL responses to protirelin across all doses compared with control men. Depressed women exhibited significant reductions in TSH responses but not in PRL responses compared with control women. CONCLUSIONS: The findings that men with major depression exhibit reductions in both protirelin-induced TSH and PRL responses support the hypothesis that TRH receptors are downregulated in depression. The findings in women are less clear and may represent the greater variance in the protirelin-induced PRL responses found in women.


Subject(s)
Depressive Disorder/blood , Prolactin/blood , Thyrotropin-Releasing Hormone/pharmacology , Thyrotropin/blood , Adult , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Female , Humans , Hydrocortisone/blood , Injections, Intravenous , Male , Middle Aged , Pituitary Gland/drug effects , Receptors, Thyrotropin-Releasing Hormone/drug effects , Sex Factors , Thyrotropin-Releasing Hormone/administration & dosage
8.
Mol Endocrinol ; 7(9): 1105-11, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8247012

ABSTRACT

Internalization of TRH receptor (TRH-R) is dependent on sequences/structures in the receptor carboxyl-terminal tail. Here, we studied whether coupling to guanine nucleotide-binding protein (G-protein) and phospholipase-C (PLC) is involved in internalization. We constructed two mutant TRH-Rs: delta 218-263 TRH-R, in which most of the residues that form the putative third intracellular loop were deleted, and D71A TRH-R, in which an Asp in the putative second transmembrane helix was mutated to Ala; these TRH-Rs did not activate PLC when expressed transiently in COS-1 cells. In contrast to wild-type (WT) TRH-Rs, approximately 60% of which were internalized at steady state after binding methyl-HisTRH, only approximately 15% of delta 218-263 and D71A TRH-Rs were internalized. Thus, mutant TRH-Rs that do not activate PLC, most likely because they are uncoupled from G-proteins, are internalized to lesser extents than WT TRH-Rs. We also studied the effects of U73122 (1-[6-[[17 beta-3-methoxyestra-1,3,5(10)-trien-17-yl]amino] hexyl]-1H-pyrrole-2,5-dione), an amino steroid that inhibits receptor-mediated activation of PLC. In COS-1 and AtT-20 cells transfected with WT TRH-Rs and in GH3 cells, U73122 virtually abolished TRH activation of PLC and partially reduced the fraction of WT TRH-Rs internalized. Thus, uncoupling WT TRH-Rs from PLC decreases internalization. We conclude that TRH-R coupling to G-protein and PLC increases the number of TRH-Rs internalized at steady state even though the primary signals for agonist-induced internalization are present in the receptor. These data support the idea that a quaternary complex of TRH/TRH-R/G protein/PLC is normally internalized.


Subject(s)
GTP-Binding Proteins/metabolism , Receptors, Thyrotropin-Releasing Hormone/metabolism , Type C Phospholipases/metabolism , Animals , Binding Sites , Cell Line , Cell Membrane/metabolism , Estrenes/pharmacology , Inositol Phosphates/metabolism , Mice , Mutagenesis, Site-Directed , Pituitary Gland/metabolism , Point Mutation , Protein Structure, Secondary , Pyrrolidinones/pharmacology , Receptors, Thyrotropin-Releasing Hormone/biosynthesis , Receptors, Thyrotropin-Releasing Hormone/drug effects , Sequence Deletion , Tetradecanoylphorbol Acetate/pharmacology , Transfection , Type C Phospholipases/antagonists & inhibitors
9.
Mol Endocrinol ; 9(11): 1455-60, 1995 Nov.
Article in English | MEDLINE | ID: mdl-8584022

ABSTRACT

A carboxyl-terminus truncated mutant of the guanine nucleotide-binding (G) protein-coupled TRH receptor (TRH-R) was previously shown to exhibit constitutive, i.e. TRH-independent, activity (C335Stop TRH-R). Chlordiazepoxide (CDE), a known competitive inhibitor of TRH binding to wild-type (WT) TRH-Rs, is shown to compete for binding to C335Stop TRH-Rs also. More importantly, CDE is shown to be a negative antagonist of C335Stop TRH-Rs. CDE rapidly caused the basal rate of inositol phosphate second messenger (IP) formation to decrease in AtT-20 pituitary cells stably expressing C335Stop TRH-Rs (AtT-C335Stop cells), but not in cells expressing WT TRH-Rs (AtT-WT cells). Similar observations were made in HeLa cells transiently expressing C335Stop or WT TRH-Rs. CDE inhibition of IP formation was shown to be specific for TRH-Rs using GH4C1 cells expressing both TRH-Rs and receptors for bombesin. In these cells, CDE inhibited TRH-stimulated IP formation, but had no effect on bombesin-stimulated IP formation. The effects of chronic administration of CDE were studied. Preincubation of AtT-C335Stop cells, but not AtT-WT cells, with CDE for several hours caused an increase in cell surface receptor number (up-regulation) that led to increased TRH stimulation of inositol phosphate formation and elevation of intracellular free Ca2+. Preincubation with CDE did not affect methyl-TRH binding affinity or TRH potency in cells expressing AtT-C335Stop or in AtT-WT cells. We conclude that CDE is a negative antagonist of C335Stop TRH-Rs and that constitutively active C335Stop TRH-Rs are down-regulated in AtT-20 pituitary cells in the absence of agonist.


Subject(s)
Chlordiazepoxide/pharmacology , Down-Regulation , Pituitary Gland, Anterior/metabolism , Receptors, Thyrotropin-Releasing Hormone/genetics , Thyrotropin-Releasing Hormone/antagonists & inhibitors , Animals , Binding, Competitive , Chlordiazepoxide/metabolism , HeLa Cells/metabolism , Humans , Inositol Phosphates/metabolism , Mice , Pituitary Neoplasms/pathology , Protein Conformation , Receptors, Bombesin/metabolism , Receptors, Thyrotropin-Releasing Hormone/biosynthesis , Receptors, Thyrotropin-Releasing Hormone/chemistry , Receptors, Thyrotropin-Releasing Hormone/drug effects , Receptors, Thyrotropin-Releasing Hormone/metabolism , Second Messenger Systems/drug effects , Thyrotropin-Releasing Hormone/metabolism , Tumor Cells, Cultured , Up-Regulation/drug effects
10.
Mol Endocrinol ; 8(8): 1038-48, 1994 Aug.
Article in English | MEDLINE | ID: mdl-7527898

ABSTRACT

TRH and GnRH receptors are each coupled to G proteins of the Gq/11 family. Activation of each of these receptors by their respective ligands results in the stimulation of phospholipase C activity, leading to calcium mobilization and protein kinase C activation. Thus, the effects of TRH and GnRH may be mediated through the same intracellular signal transduction pathway. To compare responses to TRH and GnRH directly within one cell type, we have stably transfected the rat pituitary GH3 lactotrope cell line, which expresses the endogenous TRH receptor, with an expression vector containing rat GnRH receptor cDNA. Transfected cells specifically bound GnRH with high affinity and responded to GnRH stimulation with an increase in PRL mRNA levels, analogous to their response to TRH stimulation. Stably transfected GH3 cells, which were then transiently transfected with luciferase reporter constructs containing either the PRL or the glycoprotein hormone alpha-subunit promoter, responded to either GnRH or TRH stimulation with an increase in luciferase activity in a time- and dose-dependent fashion. The stimulatory effects of maximally effective concentrations of TRH and GnRH were additive on PRL, but not alpha-subunit, gene expression. These data, coupled with evidence of cross-desensitization of alpha-subunit, but not PRL, promoter activity stimulation by TRH and GnRH, suggest that there may be differences in the signal transduction pathways activated by TRH and GnRH receptors in the regulation of PRL and alpha-subunit gene expression.


Subject(s)
Adenoma/pathology , GTP-Binding Proteins/physiology , Gene Expression Regulation/drug effects , Gonadotropin-Releasing Hormone/pharmacology , Pituitary Neoplasms/pathology , Receptors, LHRH/physiology , Receptors, Thyrotropin-Releasing Hormone/physiology , Signal Transduction , Thyrotropin-Releasing Hormone/pharmacology , Animals , Buserelin/pharmacology , Calcium/physiology , DNA, Complementary/genetics , Glycoprotein Hormones, alpha Subunit/biosynthesis , Glycoprotein Hormones, alpha Subunit/genetics , Gonadotropin-Releasing Hormone/analogs & derivatives , Luciferases/biosynthesis , Phosphatidylinositol Diacylglycerol-Lyase , Phosphoric Diester Hydrolases/physiology , Prolactin/genetics , Prolactin/metabolism , Protein Kinase C/physiology , Rats , Receptors, LHRH/drug effects , Receptors, Thyrotropin-Releasing Hormone/drug effects , Recombinant Fusion Proteins/biosynthesis , Transfection , Tumor Cells, Cultured
11.
Endocrinology ; 141(10): 3717-22, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11014227

ABSTRACT

Juxtamembrane residues in the putative third intracellular (I3) loops of a number of G protein-coupled receptors (GPCRs) have been shown to be important for coupling to G proteins. According to standard hydropathy analysis, the I3 loop of the mouse TRH receptor type 1 (mTRH-R1) is composed of 51 amino acids from position-213 to position-263. We constructed deletion and site-specific I3 loop TRH-R mutants and studied their binding and TRH-stimulated signaling activities. As expected, the effects of these mutations on TRH binding were small (less than 5-fold decreases in affinity). No effect on TRH-stimulated signaling activity was found in a mutant receptor in which the I3 loop was shortened to 16 amino acids by deleting residues from Asp-226 to Ser-260. In contrast, mutants with deletions from Asp-222 to Ser-260 or from Asp-226 to Gln-263 exhibited reduced TRH-stimulated signaling. In the region near transmembrane helix 6, single site-specific substitution of either Arg-261 or Lys-262 by neutral glutamine had little effect on signaling, but mutant TRH-Rs that were substituted by glutamine at both basic residues exhibited reduced TRH-stimulated activity. The reduced signaling activity of this doubly substituted mutant was reversed by over expressing the a subunit of Gq. These data demonstrate that the juxtamembrane regions in the TRH-R I3 loop are important for coupling to Gq.


Subject(s)
Heterotrimeric GTP-Binding Proteins/metabolism , Receptors, Thyrotropin-Releasing Hormone/genetics , Receptors, Thyrotropin-Releasing Hormone/metabolism , Amino Acid Sequence/genetics , Amino Acid Substitution , Animals , COS Cells , Female , GTP-Binding Protein alpha Subunits, Gq-G11 , Ligands , Molecular Conformation , Molecular Sequence Data , Oocytes , Receptors, Thyrotropin-Releasing Hormone/drug effects , Receptors, Thyrotropin-Releasing Hormone/physiology , Signal Transduction/drug effects , Thyrotropin-Releasing Hormone/pharmacology , Xenopus laevis
12.
FEBS Lett ; 407(3): 257-60, 1997 May 05.
Article in English | MEDLINE | ID: mdl-9175863

ABSTRACT

Wild-type and palmitoylation defective mutants of the murine G protein G11alpha were transfected into HEK293 cells. Wild-type G11alpha was membrane associated, Cys9Ser Cys10Ser G11alpha was present in the soluble fraction whilst both Cys9Ser G11alpha and Cys10Ser G11alpha were distributed between the fractions. Expression of the rat TRH receptor resulted in agonist stimulation of inositol phosphate accumulation. The degree of stimulation produced by TRH following co-transfection of the palmitoylation-resistant forms of G11alpha compared to the wild-type protein correlated with the amount of membrane-associated G protein.


Subject(s)
GTP-Binding Proteins/metabolism , Phosphoric Diester Hydrolases/metabolism , Receptors, Thyrotropin-Releasing Hormone/metabolism , Animals , Cell Line , Enzyme Activation/drug effects , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/genetics , Humans , Mice , Mutation , Palmitic Acids/chemistry , Palmitic Acids/metabolism , Rats , Receptors, Thyrotropin-Releasing Hormone/drug effects , Receptors, Thyrotropin-Releasing Hormone/genetics , Thyrotropin-Releasing Hormone/pharmacology , Transfection
13.
Neuropeptides ; 27(3): 195-202, 1994 Sep.
Article in English | MEDLINE | ID: mdl-7816190

ABSTRACT

We have recently documented the expression of preprothyrotropin-releasing hormone (TRH) gene in murine, human and rat testis. Moreover, we have localized TRH to rat Leydig cells immunohistochemically, and found that both prepro TRH mRNA and TRH levels are developmentally regulated in the rat testis. To investigate the potential roles of TRH in testicular function, characterization of TRH receptors (TRH-R) in this tissue was undertaken. Recently, a cDNA encoding murine TRH-R has been isolated, making possible cloning of a rat TRH-R cDNA from the anterior pituitary gland. This cDNA was used for detection of TRH-R gene expression in the rat testis by Northern blot analysis and reverse transcription-polymerase chain reaction (RT-PCR). TRH receptor assays were also performed with (3H)MeHisTRH as the radioactive ligand. In Northern blot analysis, a single and specific hybridization band, approximately 3.8 kb in size, was identified in whole testis RNA, identical in size with that found in the anterior pituitary gland. The concentration of TRH-R mRNA in the testis was approximately 10% of that in the pituitary. TRH-R mRNA was also detected by RT-PCR in Metrizamide gradient-purified Leydig cells. TRH receptor binding assays revealed the presence of specific, high affinity binding sites with a Kd of 1.6 x 10(-8) M in the testis. Such TRH binding was inhibited by chlordiazepoxide, a specific antagonist of TRH receptor binding. We conclude that TRH may exert local, probably autocrine, actions in the testis via a transmembrane receptor very similar or identical to that in pituitary.


Subject(s)
Receptors, Thyrotropin-Releasing Hormone/metabolism , Testis/metabolism , Animals , Base Sequence , Blotting, Northern , Cloning, Molecular , DNA Probes , GTP-Binding Proteins/metabolism , Leydig Cells/metabolism , Male , Membranes/metabolism , Molecular Sequence Data , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , RNA, Messenger/isolation & purification , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptors, Thyrotropin-Releasing Hormone/drug effects
14.
Neuropeptides ; 29(3): 171-6, 1995 Sep.
Article in English | MEDLINE | ID: mdl-8538879

ABSTRACT

The effects of single and repeated administration of amphetamine (5 mg/kg, i.p., twice a day for 14 days) on the thyrotropin-releasing hormone (TRH) level, release and receptors in the rat striatum and nucleus accumbens were evaluated. Both treatments decreased the TRH level in those structures at 2 h after the drug injection. These effects were accompanied with elevation of the basal release of TRH from the nucleus accumbens and striatal slices at the same time point, whereas the stimulated (K+, 56 mM) TRH release was attenuated following repeated amphetamine administration. Acute amphetamine had no effect on the density and affinity of TRH receptors. Repeated amphetamine increased the Bmax of TRH receptors in the striatum (by ca 49%) and nucleus accumbens (by ca 38%) at 2 h after the last drug injection. At 72 h after the last amphetamine administration, the Bmax of the TRH receptor in the striatum was still elevated (by ca 42%), whereas in the nucleus accumbens it returned to control level. No changes in the affinity of TRH receptors following repeated amphetamine were found. The obtained results indicate that repeated amphetamine evokes long- and short-term up-regulation of TRH receptors in the rat striatum and nucleus accumbens, respectively. Furthermore, it is suggested that these changes may be an adaptive response to the amphetamine-induced alterations in the TRH tissue level and release.


Subject(s)
Amphetamine/pharmacology , Corpus Striatum/drug effects , Nucleus Accumbens/drug effects , Psychomotor Performance/drug effects , Receptors, Thyrotropin-Releasing Hormone/drug effects , Thyrotropin-Releasing Hormone/metabolism , Adaptation, Physiological/drug effects , Animals , Corpus Striatum/metabolism , Drug Administration Schedule , Male , Nucleus Accumbens/metabolism , Rats , Rats, Wistar , Stimulation, Chemical
15.
Neurosci Lett ; 196(1-2): 109-12, 1995 Aug 18.
Article in English | MEDLINE | ID: mdl-7501234

ABSTRACT

We studied the mechanism by which thyrotropin-releasing hormone (TRH)-Gly stimulated prolactin and thyrotropin (TSH) secretion in pituitary, using a pituitary mammotropic cell line, GH3 cells, and a cell line stably expressing a human TRH receptor (TRH-R). In GH3 cells expressing endogenous TRH-R, an addition of TRH-Gly evoked an immediate rise of intracellular calcium concentration, indicating that TRH-Gly reacted directly without converting from TRH-Gly to TRH. In order to determine whether this reaction might occur through TRH-R, we established a cell line stably expressing a human TRH-R, by transfecting a human TRH-R cDNA into Chinese hamster ovary cells (CHO cells). In this cell line, 10 nM TRH elevated intracellular calcium significantly; the Kd for MeTRH was 1.7 nM. One micromolar and 100 nM TRH-Gly also elevated intracellular concentration of calcium significantly, but not in CHO cells which were not transfected with the TRH-R cDNA. Competition studies further revealed that TRH-Gly displaced MeTRH binding (IC50, 12 microM). These data indicate that at high concentration, TRH-Gly interacts directly with TRH-R to activate signal transduction pathway, and that release of prolactin and TSH induced by TRH-Gly in vitro may be due, at least in part, to the direct effect of TRH-Gly on the TRH-R.


Subject(s)
Pituitary Gland/drug effects , Receptors, Thyrotropin-Releasing Hormone/drug effects , Thyrotropin-Releasing Hormone/metabolism , Thyrotropin-Releasing Hormone/pharmacology , Animals , CHO Cells , Calcium/metabolism , Cell Line , Cricetinae , Dose-Response Relationship, Drug , Prolactin/metabolism , Thyrotropin/metabolism
16.
Life Sci ; 72(4-5): 601-7, 2002 Dec 20.
Article in English | MEDLINE | ID: mdl-12467901

ABSTRACT

Thyrotropin-releasing hormone (TRH) and some of its stable analogues have been shown to improve neurologic dysfunctions such as brain trauma in both animals and humans. Our previous study revealed that taltirelin, a novel orally active TRH analogue, binds to rat brain TRH receptors in vivo. The present study was undertaken to investigate whether taltirelin has neuroprotective effects in transient brain ischemia of C57BL/6J mice induced by bilateral carotid artery occlusion (2VO). Neuronal cell density in the hippocampal CA1 region of C57BL/6J mice was significantly (39.9%) decreased 1 week after 2VO-reperfusion, compared to the case of the sham group, and this reduction of hippocampal neuronal density was significantly suppressed by an intravenous (i.v.) injection of taltirelin (0.3 mg/kg). The i.v. injection of taltirelin at this dosage produced a significant increase in the dissociation constant (Kd) of specific [3H]MeTRH binding in sham and 2VO-reperfusion groups (33.6 and 51.4%, respectively) compared with the vehicle-treated group. These results indicate that the intravenously injected taltirelin bound to TRH receptors in the ischemic brain. There was little difference in the brain-to-plasma concentration ratio (Kp) of [14C]sucrose between the sham and 2VO groups of C57BL/6J mice, indicating that the tight junction of the blood-brain barrier may be intact in the ischemic brain. In conclusion, the study has shown that taltirelin may have a significant neuroprotective effect on the ischemic brain.


Subject(s)
Ischemic Attack, Transient/drug therapy , Neuroprotective Agents/therapeutic use , Nootropic Agents/therapeutic use , Receptors, Thyrotropin-Releasing Hormone/drug effects , Thyrotropin-Releasing Hormone/analogs & derivatives , Thyrotropin-Releasing Hormone/therapeutic use , Animals , Brain/pathology , Brain Chemistry/drug effects , Cell Count , Dose-Response Relationship, Drug , Hippocampus/pathology , Ischemic Attack, Transient/pathology , Male , Mice , Mice, Inbred C57BL , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , Sucrose/metabolism
17.
J Am Vet Med Assoc ; 211(1): 79-81, 1997 Jul 01.
Article in English | MEDLINE | ID: mdl-9215417

ABSTRACT

OBJECTIVE: To evaluate results of a combined dexamethasone suppression/thyrotropin-releasing hormone (TRH) stimulation test in horses suspected clinically to have a pars intermedia pituitary adenoma (PIPA). DESIGN: Case-control study. ANIMALS: 7 healthy adult horses and 5 horses suspected to have a PIPA. PROCEDURE: A baseline blood sample was collected, and dexamethasone (40 micrograms/kg [18 micrograms/lb] of body weight, IV) was administered; a second blood sample was collected 3 hours later, and TRH (1.1 mg, IV) was administered; serial blood samples were collected 15, 30, 45, 60, and 90 minutes and 21 hours after TRH administration (24 hours after dexamethasone injection). Cortisol concentration was determined for all blood samples. RESULTS: Baseline cortisol concentration was significantly lower in horses suspected to have a PIPA than in healthy horses. Cortisol concentration was suppressed by dexamethasone in both groups; however, after TRH administration, cortisol concentration returned to baseline values in horses suspected to have a PIPA, but not in healthy horses. Concentration was still less than the baseline value 24 hours after dexamethasone administration in healthy horses. CLINICAL IMPLICATIONS: The combined dexamethasone suppression/TRH stimulation test may be a useful diagnostic test in horses suspected to have a PIPA. For clinical application, collection of a blood sample 30 minutes after TRH administration is recommended.


Subject(s)
Adenoma/veterinary , Horse Diseases/physiopathology , Horses/physiology , Hydrocortisone/blood , Pituitary Neoplasms/veterinary , Adenoma/diagnosis , Adenoma/physiopathology , Animals , Case-Control Studies , Dexamethasone , Female , Glucocorticoids , Horse Diseases/diagnosis , Pituitary Gland/physiopathology , Pituitary Neoplasms/diagnosis , Pituitary Neoplasms/physiopathology , Receptors, Thyrotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Thyrotropin-Releasing Hormone/drug effects , Thyrotropin-Releasing Hormone
18.
Radiats Biol Radioecol ; 43(3): 334-6, 2003.
Article in Russian | MEDLINE | ID: mdl-12881991

ABSTRACT

The prolonged effect of thyroliberin in ULD after single intramuscular injection on contractility of lymphatic vessels directly was investigated. The controlled group of animals received injection of 0.2 ml of physiological solution. The experimental group was injected by 0.2 ml of thyroliberin in concentrations of 10(-10) or 10(-16) mol/l (1 x 10(-4) and 1 x 10(-10) micrograms/kg of the body weight respectively). During the experiment the animals were grouped in the following way: 1) directly after the injection; 2) 3 hours later; 3) on the 1st day and then every day during 2 weeks. Lymphatic vessels reactivity of the experimental animals as well as controlled was studied by application of thyroliberin and noradrenalin (in concentrations of 1 x 10(-16) and 1 x 10(-6) mol/l respectively) directly on mesentery lymphatic vessels. The lymphatic vessels reaction in control group of animals on the noradrenalin and thyroliberin was the same during the period of observation. Thyroliberin stimulated contractility at concentration of 1 x 10(-16) mol/l. The reaction of experimental group was dramatically decreased to 10(-4) mol/l on the 1st and the 3rd day (in the case i.m. injected concentration 1 x 10(-10) mol/l) and to 10(-10) mol/l (in the case of i.m. injected concentration 10(-16) mol/l). The lymphatic vessels reactivity to exogenous thyroliberin gradually established at the 6-7th days till 12th day from the moment of thyroliberin injection. The mechanisms of the action of thyroliberin in ULD are discussed.


Subject(s)
Lymphatic System/drug effects , Lymphatic System/physiology , Mesentery/drug effects , Thyrotropin-Releasing Hormone/pharmacology , Animals , Dose-Response Relationship, Drug , Endothelium, Lymphatic/drug effects , Endothelium, Lymphatic/physiology , Male , Mesentery/physiology , Muscle Contraction/drug effects , Norepinephrine/pharmacology , Rats , Receptors, Thyrotropin-Releasing Hormone/drug effects , Receptors, Thyrotropin-Releasing Hormone/metabolism
19.
Arq Bras Endocrinol Metabol ; 58(9): 962-6, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25627054

ABSTRACT

Reduced sensitivity to thyroid hormones (RSTH) is a rare disease that affects about 3,000 individuals, belonging to about 1,000 families. It results from reduced intracellular action of thyroid hormones (TH) genetically determined and manifests as persistent hyperthyroxinemia with non-suppressed thyroid-stimulating hormone (TSH). We describe a 67-years old, Caucasian woman, with past history of subtotal thyroidectomy due to diffuse goiter, who presents with a recurrence of goiter. Although she is clinically euthyroid, laboratory evaluation shows persistent hyperthyroxinemia with non-suppressed TSH. Response to thyrotropin releasing hormone (TRH) test was normal and TSH concentrations were not suppressed during oral administration of suprafisiologic doses of levothyroxine (L-T4). Peripheral blood DNA was extracted from the patient and a mutation was found localized in cluster one, at codon 346 of the ligand binding domain of the THRB gene. The patient's son underwent thyroid function testing (TFT) and genetic study, both negative, suggesting a sporadic mutation. RSTH should be considered in all hyperthyroxinemic patients who are clinically euthyroid. Mutations interfering with three major steps required for TH action on target tissues have been, so far, identified (TR-ß, TR-α, MCT8, SPB2). Each mutation is associated with a distinctive syndrome. Goal of management is to maintain a normal serum TSH level and a eumetabolic state and offer appropriate genetic counselling and prenatal diagnosis. Inappropriate treatment of eumetabolic patients results in hypothyroidism and need for TH replacement.


Subject(s)
Mutation , Rare Diseases/genetics , Thyroid Hormone Resistance Syndrome/genetics , Aged , DNA/analysis , Exons , Female , Genes, erbA , Goiter/genetics , Humans , Hyperthyroxinemia/blood , Polymerase Chain Reaction , Receptors, Thyrotropin-Releasing Hormone/blood , Receptors, Thyrotropin-Releasing Hormone/drug effects , Recurrence , Thyroid Function Tests , Thyrotropin/blood , Thyrotropin/drug effects , Thyroxine/pharmacology
20.
Menopause ; 19(11): 1253-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22713859

ABSTRACT

OBJECTIVE: Thyroid gland dysfunction is associated with menstrual cycle disturbances, infertility, and increased risk of miscarriage, but the mechanisms are poorly understood. However, little is known about the regulation of these receptors in the uterus. The aim of this study was to determine the effects of long-term treatment with steroid hormones on the expression, distribution, and regulation of the receptors for thyrotropin-releasing hormone (TRHR) and thyroid-stimulating hormone (TSHR), thyroid hormone receptor α1/α2 (THRα1/α2), and THRß1 in the uterus of surgically menopausal monkeys. METHODS: Eighty-eight cynomolgus macaques were ovariectomized and treated orally with conjugated equine estrogens (CEE; n = 20), a combination of CEE and medroxyprogesterone acetate (MPA; n = 20), or tibolone (n = 28) for 2 years. The control group (OvxC; n = 20) received no treatment. Immunohistochemistry was used to evaluate the protein expression and distribution of the receptors in luminal epithelium, glands, stroma, and myometrium of the uterus. RESULTS: Immunostaining of TRHR, TSHR, and THRs was detected in all uterine compartments. Epithelial immunostaining of TRHR was down-regulated in the CEE + MPA group, whereas in stroma, both TRHR and TSHR were increased by CEE + MPA treatment as compared with OvxC. TRHR immunoreactivity was up-regulated, but THRα and THRß were down-regulated, in the myometrium of the CEE and CEE + MPA groups. The thyroid-stimulating hormone level was higher in the CEE and tibolone groups as compared with OvxC, but the level of free thyroxin did not differ between groups. CONCLUSIONS: All receptors involved in thyroid hormone function are expressed in monkey uterus, and they are all regulated by long-term steroid hormone treatment. These findings suggest that there is a possibility of direct actions of thyroid hormones, thyroid-stimulating hormone and thyrotropin-releasing hormone on uterine function.


Subject(s)
Gonadal Steroid Hormones/pharmacology , Macaca fascicularis , Receptors, Thyroid Hormone/drug effects , Receptors, Thyrotropin-Releasing Hormone/drug effects , Receptors, Thyrotropin/drug effects , Uterus/chemistry , Animals , Estrogens, Conjugated (USP)/administration & dosage , Female , Immunohistochemistry/veterinary , Medroxyprogesterone Acetate/administration & dosage , Norpregnenes/administration & dosage , Receptors, Thyroid Hormone/analysis , Receptors, Thyrotropin/analysis , Receptors, Thyrotropin-Releasing Hormone/analysis , Thyroid Hormone Receptors alpha/analysis , Thyroid Hormone Receptors alpha/drug effects , Thyroid Hormone Receptors beta/analysis , Thyroid Hormone Receptors beta/drug effects , Uterus/physiology
SELECTION OF CITATIONS
SEARCH DETAIL