Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
J Recept Signal Transduct Res ; 38(1): 20-26, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29137494

ABSTRACT

OBJECTIVES: Extensive research has been dedicated to elucidating the mechanisms of signal transduction through different G protein-coupled receptors (GPCRs). However, relatively little is known about the regulation of receptor movement within the cell membrane upon ligand binding. In this study we focused our attention on the thyrotropin-releasing hormone (TRH) receptor that typically couples to Gq/11 proteins. METHODS: We monitored receptor diffusion in the plasma membrane of HEK293 cells stably expressing yellow fluorescent protein (YFP)-tagged TRH receptor (TRHR-YFP) by fluorescence recovery after photobleaching (FRAP). RESULTS: FRAP analysis indicated that the lateral movement of the TRH receptor was markedly reduced upon TRH binding as the value of its diffusion coefficient fell down by 55%. This effect was prevented by the addition of the TRH receptor antagonist midazolam. We also found that siRNA-mediated knockdown of Gq/11α, Gß, ß-arrestin2 and phospholipase Cß1, but not of Giα1, ß-arrestin1 or G protein-coupled receptor kinase 2, resulted in a significant decrease in the rate of TRHR-YFP diffusion, indicating the involvement of the former proteins in the regulation of TRH receptor behavior. The observed partial reduction of the TRHR-YFP mobile fraction caused by down-regulation of Giα1 and ß-arrestin1 suggests that these proteins may also play distinct roles in THR receptor-mediated signaling. CONCLUSION: These results demonstrate for the first time that not only agonist binding but also abundance of some signaling proteins may strongly affect TRH receptor dynamics in the plasma membrane.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , Receptors, Thyrotropin-Releasing Hormone/chemistry , Signal Transduction/drug effects , beta-Arrestins/chemistry , Binding Sites , Cell Membrane/drug effects , Fluorescence Recovery After Photobleaching , G-Protein-Coupled Receptor Kinase 2/chemistry , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , HEK293 Cells , Humans , Ligands , Midazolam/pharmacology , Protein Binding/drug effects , Receptors, Thyrotropin-Releasing Hormone/agonists , Receptors, Thyrotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Thyrotropin-Releasing Hormone/genetics , Signal Transduction/genetics , Thyrotropin-Releasing Hormone/chemistry , Thyrotropin-Releasing Hormone/metabolism , beta-Arrestins/genetics
2.
Nat Commun ; 8(1): 484, 2017 09 07.
Article in English | MEDLINE | ID: mdl-28883467

ABSTRACT

The hypothalamic-pituitary-thyroid (HPT) axis maintains circulating thyroid hormone levels in a narrow physiological range. As axons containing thyrotropin-releasing hormone (TRH) terminate on hypothalamic tanycytes, these specialized glial cells have been suggested to influence the activity of the HPT axis, but their exact role remained enigmatic. Here, we demonstrate that stimulation of the TRH receptor 1 increases intracellular calcium in tanycytes of the median eminence via Gαq/11 proteins. Activation of Gαq/11 pathways increases the size of tanycyte endfeet that shield pituitary vessels and induces the activity of the TRH-degrading ectoenzyme. Both mechanisms may limit the TRH release to the pituitary. Indeed, blocking TRH signaling in tanycytes by deleting Gαq/11 proteins in vivo enhances the response of the HPT axis to the chemogenetic activation of TRH neurons. In conclusion, we identify new TRH- and Gαq/11-dependent mechanisms in the median eminence by which tanycytes control the activity of the HPT axis.The hypothalamic-pituitary-thyroid (HPT) axis regulates a wide range of physiological processes. Here the authors show that hypothalamic tanycytes play a role in the homeostatic regulation of the HPT axis; activation of TRH signaling in tanycytes elevates their intracellular Ca2+ via Gαq/11 pathway, ultimately resulting in reduced TRH release into the pituitary vessels.


Subject(s)
Hypothalamo-Hypophyseal System/metabolism , Hypothalamus/cytology , Thyroid Gland/metabolism , Animals , Calcium Signaling , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Hypothalamus/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Thyrotropin-Releasing Hormone/agonists , Receptors, Thyrotropin-Releasing Hormone/genetics , Receptors, Thyrotropin-Releasing Hormone/metabolism , Thyrotropin/metabolism
3.
Eur J Pharmacol ; 761: 413-22, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26142830

ABSTRACT

Rovatirelin ([1-[-[(4S,5S)-(5-methyl-2-oxo oxazolidin-4-yl) carbonyl]-3-(thiazol-4-yl)-l-alanyl]-(2R)-2-methylpyrrolidine) is a novel synthetic agent that mimics the actions of thyrotropin-releasing hormone (TRH). The aim of this study was to investigate the electrophysiological and pharmacological effects of rovatirelin on the central noradrenergic system and to compare the results with those of another TRH mimetic agent, taltirelin, which is approved for the treatment of spinocerebellar degeneration (SCD) in Japan. Rovatirelin binds to the human TRH receptor with higher affinity (Ki=702nM) than taltirelin (Ki=3877nM). Rovatirelin increased the spontaneous firing of action potentials in the acutely isolated noradrenergic neurons of rat locus coeruleus (LC). The facilitatory action of rovatirelin on the firing rate in the LC neurons was inhibited by the TRH receptor antagonist, chlordiazepoxide. Reduction of the extracellular pH increased the spontaneous firing of LC neurons and rovatirelin failed to increase the firing frequency further, indicating an involvement of acid-sensitive K+ channels in the rovatirelin action. In in vivo studies, oral administration of rovatirelin increased both c-Fos expression in the LC and extracellular levels of noradrenaline (NA) in the medial prefrontal cortex (mPFC) of rats. Furthermore, rovatirelin increased locomotor activity. The increase in NA level and locomotor activity by rovatirelin was more potent and longer acting than those by taltirelin. These results indicate that rovatirelin exerts a central nervous system (CNS)-mediated action through the central noradrenergic system, which is more potent than taltirelin. Thus, rovatirelin may have an orally effective therapeutic potential in patients with SCD.


Subject(s)
Adrenergic Neurons/drug effects , Locus Coeruleus/drug effects , Oxazolidinones/pharmacology , Prefrontal Cortex/drug effects , Pyrrolidines/pharmacology , Thyrotropin-Releasing Hormone/pharmacology , Action Potentials , Administration, Oral , Adrenergic Neurons/metabolism , Animals , Dose-Response Relationship, Drug , Ligands , Locus Coeruleus/cytology , Locus Coeruleus/metabolism , Male , Microdialysis , Motor Activity/drug effects , Norepinephrine/metabolism , Oxazolidinones/administration & dosage , Oxazolidinones/metabolism , Prefrontal Cortex/cytology , Prefrontal Cortex/metabolism , Protein Binding , Proto-Oncogene Proteins c-fos/metabolism , Pyrrolidines/administration & dosage , Pyrrolidines/metabolism , Radioligand Assay , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Thyrotropin-Releasing Hormone/agonists , Receptors, Thyrotropin-Releasing Hormone/metabolism , Thyrotropin-Releasing Hormone/administration & dosage , Thyrotropin-Releasing Hormone/analogs & derivatives , Thyrotropin-Releasing Hormone/metabolism , Time Factors
4.
FASEB J ; 26(8): 3473-82, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22593547

ABSTRACT

G-protein-coupled receptors with dissociable agonists for thyrotropin, parathyroid hormone, and sphingosine-1-phosphate were found to signal persistently hours after agonist withdrawal. Here we show that mouse thyrotropin-releasing hormone (TRH) receptors, subtypes 2 and 1(TRH-R2 and TRH-R1), can signal persistently in HEK-EM293 cells under appropriate conditions, but TRH-R2 exhibits higher persistent signaling activity. Both receptors couple primarily to Gα(q/11). To gain insight into the mechanism of persistent signaling, we compared proximal steps of inositolmonophosphate (IP1) signaling by TRH-Rs. Persistent signaling was not caused by slower dissociation of TRH from TRH-R2 (t(1/2)=77 ± 8.1 min) compared with TRH-R1 (t(1/2)=82 ± 12 min) and was independent of internalization, as inhibition of internalization did not affect persistent signaling (115% of control), but required continuously activated receptors, as an inverse agonist decreased persistent signaling by 60%. Gα(q/11) knockdown decreased persistent signaling by TRH-R2 by 82%, and overexpression of Gα(q/11) induced persistent signaling in cells expressing TRH-R1. Lastly, persistent signaling was induced in cells expressing high levels of TRH-R1. We suggest that persistent signaling by TRHRs is exhibited when sufficient levels of agonist/receptor/G-protein complexes are established and maintained and that TRH-R2 forms and maintains these complexes more efficiently than TRH-R1.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Receptors, Thyrotropin-Releasing Hormone/physiology , Signal Transduction/physiology , Animals , Inositol Phosphates/biosynthesis , Mice , Receptors, G-Protein-Coupled , Receptors, Thyrotropin-Releasing Hormone/agonists , Receptors, Thyrotropin-Releasing Hormone/genetics
5.
ChemMedChem ; 6(3): 531-43, 2011 Mar 07.
Article in English | MEDLINE | ID: mdl-21302359

ABSTRACT

As part of our search for selective and CNS-active thyrotropin-releasing hormone (TRH) analogues, we synthesized a set of 44 new analogues in which His and pGlu residues were modified or replaced. The analogues were evaluated as agonists at TRH-R1 and TRH-R2 in cells in vitro, and in vivo in mice for analeptic and anticonvulsant activities. Several analogues bound to TRH-R1 and TRH-R2 with good to moderate affinities, and are full agonists at both receptor subtypes. Specifically, analogue 21 a (R=CH3) exhibited binding affinities (Ki values) of 0.17 µM for TRH-R1 and 0.016 µM for TRH-R2; it is 10-fold less potent than TRH in binding to TRH-R1 and equipotent with TRH in binding to TRH-R2. Compound 21 a, the most selective agonist, activated TRH-R2 with a potency (EC50 value) of 0.0021 µM, but activated TRH-R1 at EC50=0.05 µM, and exhibited 24-fold selectivity for TRH-R2 over TRH-R1. The newly synthesized TRH analogues were also evaluated in vivo to assess their potencies in antagonism of barbiturate-induced sleeping time, and several analogues displayed potent analeptic activity. Specifically, analogues 21 a,b and 22 a,b decreased sleeping time by nearly 50% more than TRH. These analogues also displayed potent anticonvulsant activity and provided significant protection against PTZ-induced seizures, but failed to provide any protection in MES-induced seizures at 10 µmol kg(-1). The results of this study provide evidence that TRH analogues that show selectivity for TRH-R2 over TRH-R1 possess potent CNS activity.


Subject(s)
Anticonvulsants/chemical synthesis , Receptors, Thyrotropin-Releasing Hormone/agonists , Thyrotropin-Releasing Hormone/analogs & derivatives , Animals , Anticonvulsants/chemistry , Anticonvulsants/therapeutic use , Disease Models, Animal , Mice , Protein Binding , Receptors, Thyrotropin-Releasing Hormone/metabolism , Seizures/chemically induced , Seizures/drug therapy , Thyrotropin-Releasing Hormone/chemical synthesis , Thyrotropin-Releasing Hormone/therapeutic use
6.
Biochem J ; 428(2): 235-45, 2010 May 13.
Article in English | MEDLINE | ID: mdl-20345371

ABSTRACT

Two GPCRs (G-protein-coupled receptors), TRHR (thyrotropin-releasing hormone receptor) and beta(2)AR (beta(2)-adrenergic receptor), are regulated in distinct manners. Following agonist binding, TRHR undergoes rapid phosphorylation attributable to GRKs (GPCR kinases); beta(2)AR is phosphorylated by both second messenger-activated PKA (protein kinase A) and GRKs with slower kinetics. TRHR co-internalizes with arrestin, whereas beta(2)AR recruits arrestin, but internalizes without it. Both receptors are dephosphorylated following agonist removal, but TRHR is dephosphorylated much more rapidly while it remains at the plasma membrane. We generated chimaeras swapping the C-terminal domains of these receptors to clarify the role of different receptor regions in phosphorylation, internalization and dephosphorylation. beta(2)AR with a TRHR cytoplasmic tail (beta(2)AR-TRHR) and TRHR with a beta(2)AR tail (TRHR-beta(2)AR) signalled to G-proteins normally. beta(2)AR-TRHR was phosphorylated well at the PKA site in the third intracellular loop, but poorly at GRK sites in the tail, whereas TRHR-beta(2)AR was phosphorylated strongly at GRK sites in the tail (Ser(355)/Ser(356) of the beta(2)AR). Both chimaeric receptors exhibited prolonged, but weak, association with arrestin at the plasma membrane, but high-affinity arrestin interactions and extensive co-internalization of receptor with arrestin required a phosphorylated TRHR tail. In contrast, swapping C-terminal domains did not change the rates of phosphorylation and dephosphorylation or the dependence of TRHR dephosphorylation on the length of agonist exposure. Thus the interactions of GPCRs with GRKs and phosphatases are determined not simply by the amino acid sequences of the substrates, but by regions outside the cytoplasmic tails.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Adrenergic beta-2 Receptor Agonists , Animals , Arrestin/metabolism , CHO Cells , Cell Line , Cell Membrane/metabolism , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme-Linked Immunosorbent Assay , G-Protein-Coupled Receptor Kinases/metabolism , Humans , Inositol Phosphates/metabolism , Isoproterenol/pharmacology , Microscopy, Fluorescence , Phosphorylation/drug effects , Protein Binding , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics , Receptors, Thyrotropin-Releasing Hormone/agonists , Receptors, Thyrotropin-Releasing Hormone/genetics , Receptors, Thyrotropin-Releasing Hormone/metabolism , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology
7.
J Neurosci ; 29(14): 4471-83, 2009 Apr 08.
Article in English | MEDLINE | ID: mdl-19357273

ABSTRACT

The histaminergic tuberomamillary nucleus (TMN) controls arousal and attention, and the firing of TMN neurons is state-dependent, active during waking, silent during sleep. Thyrotropin-releasing hormone (TRH) promotes arousal and combats sleepiness associated with narcolepsy. Single-cell reverse-transcription-PCR demonstrated variable expression of the two known TRH receptors in the majority of TMN neurons. TRH increased the firing rate of most (ca 70%) TMN neurons. This excitation was abolished by the TRH receptor antagonist chlordiazepoxide (CDZ; 50 mum). In the presence of tetrodotoxin (TTX), TRH depolarized TMN neurons without obvious change of their input resistance. This effect reversed at the potential typical for nonselective cation channels. The potassium channel blockers barium and cesium did not influence the TRH-induced depolarization. TRH effects were antagonized by inhibitors of the Na(+)/Ca(2+) exchanger, KB-R7943 and benzamil. The frequency of GABAergic spontaneous IPSCs was either increased (TTX-insensitive) or decreased [TTX-sensitive spontaneous IPSCs (sIPSCs)] by TRH, indicating a heterogeneous modulation of GABAergic inputs by TRH. Facilitation but not depression of sIPSC frequency by TRH was missing in the presence of the kappa-opioid receptor antagonist nor-binaltorphimine. Montirelin (TRH analog, 1 mg/kg, i.p.) induced waking in wild-type mice but not in histidine decarboxylase knock-out mice lacking histamine. Inhibition of histamine synthesis by (S)-alpha-fluoromethylhistidine blocked the arousal effect of montirelin in wild-type mice. We conclude that direct receptor-mediated excitation of rodent TMN neurons by TRH demands activation of nonselective cation channels as well as electrogenic Na(+)/Ca(2+) exchange. Our findings indicate a key role of the brain histamine system in TRH-induced arousal.


Subject(s)
Histamine/physiology , Hypothalamic Area, Lateral/physiology , Neurons/physiology , Thyrotropin-Releasing Hormone/physiology , Action Potentials/physiology , Animals , Calcium Channels/metabolism , Calcium Channels/physiology , Cations, Divalent/metabolism , Histamine/deficiency , Hypothalamic Area, Lateral/metabolism , Inhibitory Postsynaptic Potentials/physiology , Male , Mice , Mice, Knockout , Neurons/metabolism , Rats , Rats, Wistar , Receptors, Thyrotropin-Releasing Hormone/agonists , Receptors, Thyrotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Thyrotropin-Releasing Hormone/physiology , Sleep Stages/physiology , Sodium Channels/metabolism , Sodium Channels/physiology
8.
J Mol Graph Model ; 27(3): 309-20, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18595758

ABSTRACT

Design and development of therapeutically useful CNS selective thyrotropin-releasing hormone (TRH) analogs acting on TRH-R2 receptor subtype, exerting weak or no TRH-R1-mediated TSH-releasing side effects has gained imagination of researchers in the recent past. The present study reports the development and implementation of a selectivity-based QSAR approach for screening selective agonists of TRH-R2 receptor subtype. The statistically significant predictive models were thoroughly validated using an external validation set whose activity was previously unknown. The model was able to predict preference for either of the receptor subtypes successfully.


Subject(s)
Drug Design , Drug Evaluation , Quantitative Structure-Activity Relationship , Receptors, Thyrotropin-Releasing Hormone/agonists , Thyrotropin-Releasing Hormone/analogs & derivatives , Models, Molecular , Receptors, Thyrotropin-Releasing Hormone/chemistry , Reproducibility of Results
9.
J Nat Prod ; 71(5): 884-6, 2008 May.
Article in English | MEDLINE | ID: mdl-18407692

ABSTRACT

High-throughput screening of a plant and marine invertebrate extract library to find natural products with rat thyrotropin releasing hormorne (TRH) receptor 2 binding affinity led to the isolation of four new (1-4) and one known (5) spongian diterpene from the sponge Spongia sp. The structures were assigned from interpretation of 2D NMR and high-resolution ESIMS data. The absolute configurations of 1-4 were proposed on the basis of analysis of their CD spectra. Diterpenes 1-5 showed rat TRH receptor 2 binding affinity with IC(50) values of 23 microM, 70 microM, 400 microM, 600 microM, and 1 mM, respectively.


Subject(s)
Diterpenes/isolation & purification , Diterpenes/pharmacology , Porifera/chemistry , Receptors, Thyrotropin-Releasing Hormone/agonists , Animals , Diterpenes/chemistry , Inhibitory Concentration 50 , Molecular Structure , Oceans and Seas , Rats
10.
J Nat Prod ; 71(5): 881-3, 2008 May.
Article in English | MEDLINE | ID: mdl-18412395

ABSTRACT

High-throughput screeing of a plant and marine invertebrate extract library to find natural products with rat thytotropin releasing hormone receptor 2 binding affinity led to the isolation of two new active acylphloroglucinols, corymbones A and B (1 and 2) from flowers of the Queensland tree Corymbia peltata. Their structures were assigned from interpretation of 2D NMR and high-resolution ESIMS data. Compounds 1 and 2 showed rat TRH receptor 2 binding affinity with IC 50 values of 23 and 19 microM, respectively.


Subject(s)
Myrtaceae/chemistry , Phloroglucinol/analogs & derivatives , Phloroglucinol/isolation & purification , Phloroglucinol/pharmacology , Plants, Medicinal/chemistry , Receptors, Thyrotropin-Releasing Hormone/agonists , Animals , Flowers/chemistry , Nuclear Magnetic Resonance, Biomolecular , Phloroglucinol/chemistry , Queensland , Rats
11.
Physiol Res ; 57(2): 195-203, 2008.
Article in English | MEDLINE | ID: mdl-17552882

ABSTRACT

Prolonged agonist stimulation results in specific transfer of activated Galpha subunits of G(q)alpha/G(11)alpha family from particulate membrane fraction to soluble (cytosol) cell fraction isolated as 250,000 x g supernatant. In this study, we have used 2D electrophoresis for more defined resolution of Galpha subunits of G(q)alpha/G(11)alpha family and followed the time course of solubilization effect. The small signal of soluble G proteins was already detected in control, hormone-unexposed cells. Hormone stimulation resulted in a slow but continuous increase of both intensity and number of immunoreactive signals/spots of these G proteins (10, 30, 60, 120 and 240 min). At longer times of agonist exposure (>2 hours), a marked increase of G(q)alpha/G(11)alpha proteins was detected. The maximal level of soluble G(q)alpha/G(11)alpha proteins was reached after 16 hours of continuous agonist exposure. At this time interval, eight individual immunoreactive signals of G(q)alpha/G(11)alpha proteins could be resolved. The relative proportion among these spots was 15:42:10:11:7:7:2:5. Solubilization of this class of Galpha proteins was thus observed after prolonged agonist stimulation only, induced by ultra high concentration of hormone and in cells expressing a large number of GPCRs. Our data therefore rather indicate tight/persisting binding of G(q)alpha/G(11)alpha proteins to the membrane.


Subject(s)
Cell Membrane/metabolism , GTP-Binding Protein alpha Subunits/metabolism , Protein Transport/physiology , Receptors, Thyrotropin-Releasing Hormone/metabolism , Second Messenger Systems/physiology , Animals , Cell Line , Cell Membrane/drug effects , Cytosol/drug effects , Cytosol/metabolism , Electrophoresis, Gel, Two-Dimensional , GTP-Binding Protein alpha Subunits/drug effects , Humans , Mice , Protein Transport/drug effects , Rats , Receptors, Thyrotropin-Releasing Hormone/agonists , Second Messenger Systems/drug effects , Solubility , Time Factors , Transfection
12.
J Neurochem ; 103(3): 1102-10, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17760865

ABSTRACT

We examined the involvement of thyrotropin-releasing hormone (TRH) and TRH type 1 and 2 receptors (TRH-R1 and TRH-R2, respectively) in the regulation of hypothalamic neuronal histamine. Infusion of 100 nmol TRH into the rat third cerebroventricle (3vt) significantly decreased food intake (p < 0.05) compared to controls infused with phosphate- buffered saline. This TRH-induced suppression of food intake was attenuated partially in histamine-depleted rats pre-treated with alpha-fluoromethylhistidine (a specific suicide inhibitor of histidine decarboxylase) and in mice with targeted disruption of histamine H1 receptors. Infusion of TRH into the 3vt increased histamine turnover as assessed by pargyline-induced accumulation of tele-methylhistamine (t-MH, a major metabolite of neuronal histamine in the brain) in the tuberomammillary nucleus (TMN), the paraventricular nucleus, and the ventromedial hypothalamic nucleus in rats. In addition, TRH-induced decrease of food intake and increase of histamine turnover were in a dose-dependent manner. Microinfusion of TRH into the TMN increased t-MH content, histidine decarboxylase (HDC) activity and expression of HDC mRNA in the TMN. Immunohistochemical analysis revealed that TRH-R2, but not TRH-R1, was expressed within the cell bodies of histaminergic neurons in the TMN of rats. These results indicate that hypothalamic neuronal histamine mediates the TRH-induced suppression of feeding behavior.


Subject(s)
Appetite Regulation/physiology , Histamine/metabolism , Hypothalamus/metabolism , Neurons/metabolism , Receptors, Thyrotropin-Releasing Hormone/metabolism , Thyrotropin-Releasing Hormone/metabolism , Animals , Appetite Regulation/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Histamine H1 Antagonists/pharmacology , Histidine Decarboxylase/antagonists & inhibitors , Histidine Decarboxylase/genetics , Histidine Decarboxylase/metabolism , Hypothalamus/anatomy & histology , Hypothalamus/drug effects , Immunohistochemistry , Injections, Intraventricular , Male , Methylhistamines/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Receptors, Histamine H1/drug effects , Receptors, Histamine H1/genetics , Receptors, Histamine H1/metabolism , Receptors, Thyrotropin-Releasing Hormone/agonists , Thyrotropin-Releasing Hormone/pharmacology
13.
Bioorg Med Chem ; 15(1): 433-43, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17035026

ABSTRACT

Thyrotropin-releasing hormone (TRH) analogs in which the N-1(tau) or the C-2 position of the imidazole ring of the histidine residue is substituted with various alkyl groups and the l-pyroglutamic acid (pGlu) is replaced with the l-pyro-2-aminoadipic acid (pAad) or (R)- and (S)-3-oxocyclopentane-1-carboxylic acid (Ocp) were synthesized and studied as agonists for TRH receptor subtype 1 (TRH-R1) and subtype 2 (TRH-R2). We observed that several analogs were selective agonists of TRH-R2 showing relatively less or no activation of TRH-R1. For example, the most selective agonist of the series 13, in which pGlu is replaced with the pAad and histidine residue is substituted at the N-1 position with an isopropyl group, was found to activate TRH-R2 with a potency (EC(50)=1.9microM) but did not activate TRH-R1 (potency>100 microM); that is, exhibited >51-fold greater selectivity for TRH-R2 versus TRH-R1. Analog 8, in which pGlu is replaced with pAad and histidine is substituted at the N-1(tau) position with a methyl group, exhibited a binding affinity (K(i)=0.0032 microM) to TRH-R1 that is similar to that of [Ntau(1)-Me-His]-TRH and displayed potent activation of TRH-R1 and TRH-R2 (EC(50)=0.0049 and 0.0024 microM, respectively). None of the analogs in which pGlu is replaced with the bioisosteric (R)- and (S)-(Ocp) and the imidazole ring is substituted at the N-1(tau) or C-2 position were found to bind or activate either TRH-R1 or TRH-R2 at the highest test dose of 100 microM.


Subject(s)
Histidine/chemistry , Pyrrolidonecarboxylic Acid/chemistry , Receptors, Thyrotropin-Releasing Hormone/agonists , Thyrotropin-Releasing Hormone/chemical synthesis , Thyrotropin-Releasing Hormone/pharmacology , Molecular Conformation , Protein Binding , Receptors, Thyrotropin-Releasing Hormone/chemistry , Stereoisomerism , Structure-Activity Relationship , Thyrotropin-Releasing Hormone/analogs & derivatives
14.
J Biol Chem ; 281(19): 13103-13109, 2006 May 12.
Article in English | MEDLINE | ID: mdl-16551618

ABSTRACT

We show that several analogs of thyrotropin-releasing hormone (TRH) are more efficacious agonists at TRH receptors R1 and R2 than TRH itself. The apparent efficacies of the analogs were inversely related to their potencies and were independent of the nature of the modifications in TRH structure. In studies in intact cells, we showed that the differences in apparent efficacies were not due to differences in G-protein coupling, receptor desensitization, or recycling. Moreover, the differences in efficacies persisted in experiments using accessory protein-free membranes. We conclude that the efficacy differences of TRH analogs originated from the enhanced ability of TRH-R complexed to the low affinity agonists to directly activate G-protein(s), and not by a modulation of the activity of accessory proteins, and propose possible mechanisms for this phenomenon.


Subject(s)
Receptors, Thyrotropin-Releasing Hormone/agonists , Thyrotropin-Releasing Hormone/analogs & derivatives , Thyrotropin-Releasing Hormone/pharmacology , Cell Line , Dose-Response Relationship, Drug , GTP-Binding Proteins/metabolism , Gene Expression Regulation , Humans , Molecular Structure , Protein Binding , Thyrotropin-Releasing Hormone/chemistry
15.
J Med Chem ; 48(19): 6162-5, 2005 Sep 22.
Article in English | MEDLINE | ID: mdl-16162016

ABSTRACT

Thyrotropin-releasing hormone (TRH) analogues in which the C-2 position of the imidazole ring of the centrally placed histidine residue is substituted with various alkyl groups were synthesized and studied as agonists for TRH receptor subtype 1 (TRH-R1) and subtype 2 (TRH-R2). Several analogues were found to be selective agonists for TRH-R2 exhibiting no activation of TRH-R1. For example, analogue 4 (R= c-C3H5) was found to activate TRH-R2 with a potency (EC50) of 0.41 microM but did not activate TRH-R1 (potency > 100 microM). This study describes the first discovery of TRH-R2-specific agonists and provides impetus to design predominately CNS-effective TRH peptides.


Subject(s)
Central Nervous System Agents/chemical synthesis , Receptors, Thyrotropin-Releasing Hormone/agonists , Thyrotropin-Releasing Hormone/analogs & derivatives , Thyrotropin-Releasing Hormone/chemical synthesis , Cell Line , Central Nervous System Agents/chemistry , Central Nervous System Agents/pharmacology , Humans , Imidazoles/chemistry , Structure-Activity Relationship , Thyrotropin-Releasing Hormone/pharmacology
16.
Biochemistry ; 44(7): 2419-31, 2005 Feb 22.
Article in English | MEDLINE | ID: mdl-15709754

ABSTRACT

The conformational changes at the cytoplasmic ends of transmembrane helices 5 and 6 (TMH5 and TMH6) of thyrotropin-releasing hormone (TRH) receptor type I (TRH-R1) during activation were analyzed by cysteine-scanning mutagenesis followed by disulfide cross-linking and molecular modeling. Sixteen double cysteine mutants were constructed by substitution of one residue at the cytoplasmic end of TMH5 and the other at that of TMH6. The cross-linking experiments indicate that four mutants, Q263C/G212C, Q263C/Y211C, T265C/G212C, and T265C/Y211C, exhibited disulfide bond formation that was sensitive to TRH occupancy. We refined our previous TRH-R1 models by embedding them into a hydrated explicit lipid bilayer. Molecular dynamics simulations of the models, as well as in silico double cysteine models, generated trajectories that were in agreement with experimental results. Our findings suggest that TRH binding induces a separation of the cytoplasmic ends of TMH5 and TMH6 and a rotation of TMH6. These changes likely increase the surface accessible area at the juxtamembrane region of intracellular loop 3 that could promote interactions between G proteins and key residues within the receptor.


Subject(s)
Cross-Linking Reagents/chemistry , Disulfides/chemistry , Models, Molecular , Receptors, Thyrotropin-Releasing Hormone/agonists , Receptors, Thyrotropin-Releasing Hormone/chemistry , Thyrotropin-Releasing Hormone/chemistry , Amino Acid Sequence , Animals , Cell Line , Computational Biology/methods , Computer Simulation , Cysteine/chemistry , Cysteine/genetics , Cytoplasm/chemistry , Cytoplasm/genetics , Cytoplasm/metabolism , Humans , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Phenanthrolines/chemistry , Protein Conformation , Protein Structure, Secondary/genetics , Receptors, Thyrotropin-Releasing Hormone/genetics , Thyrotropin-Releasing Hormone/metabolism
17.
Biochem J ; 380(Pt 3): 815-21, 2004 Jun 15.
Article in English | MEDLINE | ID: mdl-15025564

ABSTRACT

To study the effect of agonist on the TRH (thyrotrophin-releasing hormone) receptor protein, an epitope-tagged receptor was stably expressed in HEK-293 cells (human embryonic kidney 293 cells) and receptor levels were measured by immunoblotting. TRH caused a 5-25-fold increase in receptor protein during 48 h, which was half-maximal at 1 nM and was slowly reversible after hormone withdrawal. Chlordiazepoxide, an inverse agonist, had no effect. TRH up-regulation was mimicked by phorbol ester and blocked by the protein kinase C inhibitor GF109203X in combination with thapsigargin, which prevents a calcium response. TRH and phorbol ester increased the density of immunoreactive receptors localized at the cell surface and [3H]MeTRH (where MeTRH stands for [N3-methyl-His]TRH) binding. TRH also increased the concentration of a truncated, internalization-defective receptor. Analysis of cell lines stably expressing TRH receptors fused to the green fluorescent protein on a fluorescence-activated cell sorter showed that TRH and phorbol ester caused 2.7- and 6.8-fold increases in fusion protein expression respectively. TRH receptor up-regulation was only partially accounted for by changes in receptor mRNA, which increased 1.7-fold. TRH caused a small increase in receptor concentration in the presence of cycloheximide, actinomycin D or MG132. In contrast with the results obtained with the TRH receptor, agonist decreased the concentration of stably expressed b2-adrenergic receptors. These results show that TRH increases receptor concentration by a complex mechanism that requires signal transduction but not receptor endocytosis.


Subject(s)
Receptors, Thyrotropin-Releasing Hormone/agonists , Receptors, Thyrotropin-Releasing Hormone/metabolism , Up-Regulation/physiology , Cell Line , Humans , Kidney/chemistry , Kidney/cytology , Kidney/embryology
18.
Biochem J ; 372(Pt 3): 851-9, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12628004

ABSTRACT

Thyrotropin-releasing hormone (TRH) receptor (TRHR) is a G-protein-coupled receptor playing a crucial role in the anterior pituitary where it controls the synthesis and secretion of thyroid-stimulating hormone and prolactin. Its widespread presence not only in the central nervous system, but also in peripheral tissues, including thymus, indicates other important, but unknown, functions. One hypothesis is that the neuropeptide TRH could play a role in the immune system. We report here that the human TRHR promoter contains 11 putative response elements for the haematopoietic transcription factor c-Myb and is highly Myb-responsive in transfection assays. Analysis of Myb binding to putative response elements revealed one preferred binding site in intron 1 of the receptor gene. Transfection studies of promoter deletions confirmed that this high-affinity element is necessary for efficient Myb-dependent transactivation of reporter plasmids in CV-1 cells. The Myb-dependent activation of the TRHR promoter was strongly suppressed by expression of a dominant negative Myb-Engrailed fusion. In line with these observations, reverse transcriptase PCR analysis of rat tissues showed that the TRHR gene is expressed both in thymocytes and bone marrow. Furthermore, specific, high-affinity TRH agonist binding to cell-surface receptors was demonstrated in thymocytes and a haematopoietic cell line. Our findings imply a novel functional link between the neuroendocrine and the immune systems at the level of promoter regulation.


Subject(s)
Hematopoietic Stem Cells/physiology , Proto-Oncogene Proteins c-myb/physiology , Receptors, Thyrotropin-Releasing Hormone/genetics , Animals , Binding Sites , COS Cells , Cell Line , Gene Expression Regulation/physiology , HeLa Cells , Hematopoietic Stem Cells/metabolism , Humans , Introns , Neurosecretory Systems/metabolism , Neurosecretory Systems/physiology , Plasmids/genetics , Plasmids/metabolism , Promoter Regions, Genetic/physiology , Protein Binding , Proto-Oncogene Proteins c-myb/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Receptors, Thyrotropin-Releasing Hormone/agonists , Receptors, Thyrotropin-Releasing Hormone/metabolism , Response Elements/physiology , Thymus Gland/cytology , Thymus Gland/metabolism , Transcriptional Activation , Transfection
19.
Biotechniques ; 33(5): 1152-4, 1156-7, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12449397

ABSTRACT

G protein-coupled receptors (GPCRs) are the largest family of proteins involved in transmembrane signal transduction and are actively studied because of their suitability as therapeutic small-molecule drug targets. Agonist activation of GPCRs almost invariably results in the receptor being desensitized. One of the key events in receptor desensitization is the sequestration of the receptor from the cell surface into acidic intracellular endosomes. Therefore, a convenient, generic, and noninvasive monitor of this process is desirable. A novel, pH-sensitive, red-excited fluorescent dye, CypHer 5, was synthesized. This dye is non-fluorescent at neutral pH and is fluorescent at acidic pH. Anti-epitope antibodies labeled with this dye were internalized in an agonist concentration- and time-dependent manner, following binding on live cells to a range of GPCRs that had been modified to incorporate the epitope tags in their extracellular N-terminal domain. This resulted in a large signal increase over background. When protonated, the red fluorescence of CypHer 5 provides a generic reagent suitable for monitoring the internalization of GPCRs into acidic vesicles. This approach should be amenable to the study of many other classes of cell surface receptors that also internalize following stimulation.


Subject(s)
Carbocyanines/analysis , Endocytosis/drug effects , Fluorescent Dyes/analysis , GTP-Binding Proteins/physiology , Receptors, Thyrotropin-Releasing Hormone/agonists , Thyrotropin/pharmacology , Animals , Antibodies, Anti-Idiotypic/immunology , Antigen-Antibody Reactions , CHO Cells , Calcium Signaling , Cell Line , Cricetinae , Cricetulus , Endosomes/chemistry , Enkephalin, Leucine-2-Alanine/pharmacology , Epitopes/immunology , Genes, Reporter , Green Fluorescent Proteins , Humans , Hydrogen-Ion Concentration , Iloprost/pharmacology , Kidney , Luminescent Proteins/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Protein Structure, Tertiary , Receptors, Epoprostenol , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/genetics , Receptors, Prostaglandin/agonists , Receptors, Prostaglandin/genetics , Receptors, Thyrotropin-Releasing Hormone/genetics , Recombinant Fusion Proteins/agonists , Transfection , Vesicular stomatitis Indiana virus/genetics , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
20.
Eur J Endocrinol ; 147(3): 363-9, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12213674

ABSTRACT

OBJECTIVE: Thyrotropin-releasing hormone (TRH) is inactivated in the extracellular compartment by pyroglutamyl aminopeptidase II (PPII), a narrow specificity ectopeptidase present in the brain and in the lactotrophs of the adenohypophysis. TRH and various hypothalamic/paracrine agents regulate the activity of PPII on the surface of adenohypophyseal cells in primary culture. The activity of the hypothalamic-pituitary-thyroid axis presents circadian variations including an increase of serum thyrotropin levels in the early hours of the day. The purpose of this study was to determine whether adenohypophyseal PPII activity fluctuates during the daytime in the male rat and the role of TRH in these regulatory events in vivo. RESULTS: Adenohypophyseal PPII specific activity and mRNA levels presented diurnal variations. A decrease in specific activity occurred with a minimum between 0930 and 1130 h, associated with increased serum thyrotropin levels. PPII mRNA levels were lowest at 0800 h. Intraperitoneal injection at 0800 or 1000 h of [3-Me-His(2)]-TRH, a potent agonist of the TRH receptor, reduced PPII specific activity at 30 min post-injection which was followed by a return to basal levels at 2 h. A second phase of decrease occurred between 4 and 8 h post-injection. Intravenous injection of a TRH-immune serum induced, at 2 h post-injection, an increase in adenohypophyseal PPII specific activity, which lasted up to 6 h. CONCLUSIONS: Adenohypophyseal PPII activity and mRNA levels fluctuate during the day; TRH down-regulates PPII activity in vivo, contributing to some of these variations. These new findings, and previous data, suggest that adenohypophyseal PPII activity varies in distinct physiological events, in response to endocrine and hypothalamic/paracrine factors, potentially modulating responses to TRH.


Subject(s)
Aminopeptidases/metabolism , Circadian Rhythm , Pituitary Gland, Anterior/enzymology , Thyrotropin-Releasing Hormone/analogs & derivatives , Thyrotropin-Releasing Hormone/pharmacology , Aminopeptidases/genetics , Animals , Gene Expression Regulation, Enzymologic/drug effects , Immune Sera/administration & dosage , Injections, Intravenous , Male , Pyrrolidonecarboxylic Acid/analogs & derivatives , RNA, Messenger/analysis , Rats , Rats, Wistar , Receptors, Thyrotropin-Releasing Hormone/agonists , Thyrotropin/blood , Thyrotropin-Releasing Hormone/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...