Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
2.
Poult Sci ; 99(3): 1643-1654, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32115036

ABSTRACT

The physiological roles of thyrotropin-releasing hormone (TRH) are proposed to be mediated by TRH receptors (TRHR), which have been divided into 3 subtypes, namely, TRHR1, TRHR2, and TRHR3, in vertebrates. Although 2 TRH receptors (TRHR1 and TRHR3) have been predicted to exist in birds, it remains unclear whether TRHR3 is a functional TRH receptor similar to TRHR1. Here, we reported the functionality and tissue expression of TRHR3 in chickens. The cloned chicken TRHR3 (cTRHR3) encodes a receptor of 387 amino acids, which shares high-amino-acid identities (63-80%) to TRHR3 of parrots, lizards, Xenopus tropicalis, and tilapia and comparatively lower sequence identities to chicken TRHR1 or mouse TRHR2. Using cell-based luciferase reporter assays and Western blot, we demonstrated that similar to chicken TRHR1 (cTRHR1), cTRHR3 expressed in HEK 293 cells can be potently activated by TRH and that its activation stimulates multiple signaling pathways, indicating both TRH receptors are functional. Quantitative real-time PCR revealed that cTRHR1 and cTRHR3 are widely, but differentially, expressed in chicken tissues, and their expression is likely controlled by promoters located upstream of exon 1, which display strong promoter activities in cultured DF-1 cells. cTRHR1 is highly expressed in the anterior pituitary and testes, while cTRHR3 is highly expressed in the muscle, testes, fat, pituitary, spinal cord, and many brain regions (including hypothalamus). These findings indicate that TRH actions are likely mediated by 2 TRH receptors in chickens. In conclusion, our data provide the first piece of evidence that both cTRHR3 and cTRHR1 are functional TRH receptors, which helps to elucidate the physiological roles of TRH in birds.


Subject(s)
Amino Acid Sequence , Chickens/genetics , Receptors, Thyrotropin-Releasing Hormone/genetics , Animals , Cell Line , Cloning, Molecular , Ducks , Female , Gene Expression , HEK293 Cells , Humans , Male , Real-Time Polymerase Chain Reaction/veterinary , Receptors, Thyrotropin-Releasing Hormone/chemistry , Receptors, Thyrotropin-Releasing Hormone/metabolism , Thyrotropin-Releasing Hormone/metabolism
3.
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
4.
Eur J Med Chem ; 111: 72-83, 2016 Mar 23.
Article in English | MEDLINE | ID: mdl-26854379

ABSTRACT

Thyrotropin-releasing hormone (TRH) analogues bearing halogen groups (Cl, Br and I) at the C-2 and/or C-5 position, and the alkyl group (CH3, C2H5, C3H7, CH2C6H5) at the N-1 position of the imidazole ring of the central histidine residue were synthesized and evaluated for the receptor binding, calcium mobilization (FLIPR), and IP-1 assay at the HEK mTRHR1 and HEK mTRHR2 expressing cell lines. The most promising analogue 7k showed 925-fold selectivity for HEK mTRH-R2 receptor subtype in the IP-1 assay, 272-fold selectivity for HEK mTRH-R2 receptor subtype in the FLIPR assay, and 21-fold receptor binding specificity at HEK TRH-R2 receptor subtype. The peptide 7k was evaluated in vitro in a brain membrane competitive binding assay, and for stability analysis in the presence of TRH-DE, in vivo. The analogue 7k showed decrease in the sleeping time by more than 76% in a pentobarbital-induced sleeping assay, and showed comparatively less elevation in the TSH level in the blood, in vivo. The computational homology modeling of TRH-R1 and TRH-R2 and docking study with the most potent peptide 7k provide impetus to design CNS specific TRH analogues.


Subject(s)
Histidine/metabolism , Thyrotropin-Releasing Hormone/analogs & derivatives , Thyrotropin-Releasing Hormone/metabolism , HEK293 Cells , Histidine/chemistry , Humans , Models, Molecular , Molecular Conformation , Receptors, Thyrotropin-Releasing Hormone/chemistry , Receptors, Thyrotropin-Releasing Hormone/metabolism , Thyrotropin-Releasing Hormone/chemistry
5.
Bioorg Med Chem ; 23(17): 5641-53, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26216015

ABSTRACT

Thyrotropin-releasing hormone (TRH)-like peptides were synthesized by replacing critical histidine and pGlu residues in the native peptide. The peptides were evaluated in vitro for receptor binding activity assay and in the cell functional assay; the peptides exhibit selective basal signaling agonist behavior toward TRH-R2. For example, peptides 8a, 8b, 8c, 8 f, 8 h, 8 l and 12 d activated TRH-R2 with potency (EC50) of 0.53 µM, 0.048 µM, 0.05 µM, 0.006 µM, 0.31 µM, 0.034 µM and 0.004 µM, respectively. In contrast for signaling activation of TRH-R1, the same peptide required higher concentration of 19.35 µM, 3.98 µM, 2.54 µM, 0.287 µM, 11.28 µM, 0.986 µM and 0.944 µM, respectively. The results showed that peptides were 36.5, 82.9, 50.8, 47.8, 36.3, 32.6 and 235-fold selective to TRH-R2 receptor subtype. The peptides were investigated for CNS activity at 10 µmol/kg in pentobarbital-induced sleep assay study. Peptides 8c (16.5 ± 1.4 min) and 8l (16.5 ± 2.1 min) displayed excellent CNS activity. In an in vivo study, peptide 8c did not cause significant change in the rat plasma TSH levels. The peptide 8c was further investigated for neuroprotective potential, and significantly reduced infracts volume and neurological score in the focal cerebral ischemia model in mice. Peptide 8c also significantly lowered MDA levels, indicating reduction of oxidative and enhanced percentage cell survival in CA1 region, when compared to ischemic brain.


Subject(s)
Cognition Disorders/genetics , Peptides/metabolism , Receptors, Thyrotropin-Releasing Hormone/chemistry , Thyrotropin-Releasing Hormone/chemical synthesis , Animals , Brain Ischemia , Mice , Molecular Structure , Rats
6.
Biochim Biophys Acta ; 1848(3): 781-96, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25485475

ABSTRACT

UNLABELLED: Here we investigated the effect of disruption of plasma membrane integrity by cholesterol depletion on thyrotropin-releasing hormone receptor (TRH-R) surface mobility in HEK293 cells stably expressing TRH-R-eGFP fusion protein (VTGP cells). Detailed analysis by fluorescence recovery after photobleaching (FRAP) in bleached spots of different sizes indicated that cholesterol depletion did not result in statistically significant alteration of mobile fraction of receptor molecules (Mf). The apparent diffusion coefficient (Dapp) was decreased, but this decrease was detectable only under the special conditions of screening and calculation of FRAP data. Analysis of mobility of receptor molecules by raster image correlation spectroscopy (RICS) did not indicate any significant difference between control and cholesterol-depleted cells. Results of our FRAP and RICS experiments may be collectively interpreted in terms of a "membrane fence" model which regards the plasma membrane of living cells as compartmentalized plane where lateral diffusion of membrane proteins is limited to restricted areas by cytoskeleton constraints. Hydrophobic interior of plasma membrane, studied by steady-state and time-resolved fluorescence anisotropy of hydrophobic membrane probe DPH, became substantially more "fluid" and chaotically organized in cholesterol-depleted cells. Decrease of cholesterol level impaired the functional coupling between the receptor and the cognate G proteins of Gq/G11 family. IN CONCLUSION: the presence of an unaltered level of cholesterol in the plasma membrane represents an obligatory condition for an optimum functioning of TRH-R signaling cascade. The decreased order and increased fluidity of hydrophobic membrane interior suggest an important role of this membrane area in TRH-R-Gq/G11α protein coupling.


Subject(s)
Cell Membrane/metabolism , Cholesterol/metabolism , Green Fluorescent Proteins/metabolism , Receptors, Thyrotropin-Releasing Hormone/metabolism , Algorithms , Cell Membrane/chemistry , Diffusion , Diphenylhexatriene/chemistry , Diphenylhexatriene/metabolism , Fluorescence Polarization , Fluorescence Recovery After Photobleaching , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , HEK293 Cells , Humans , Kinetics , Microscopy, Confocal , Protein Binding , Protein Transport , Receptors, Thyrotropin-Releasing Hormone/chemistry , Receptors, Thyrotropin-Releasing Hormone/genetics
7.
Cell Struct Funct ; 37(1): 1-12, 2012.
Article in English | MEDLINE | ID: mdl-22240728

ABSTRACT

Protein-protein interactions define specificity in signal transduction and these interactions are central to transmembrane signaling by G-protein-coupled receptors (GPCRs). It is not quite clear, however, whether GPCRs and the regulatory trimeric G-proteins behave as freely and independently diffusible molecules in the plasma membrane or whether they form some preassociated complexes. Here we used clear-native polyacrylamide gel electrophoresis (CN-PAGE) to investigate the presumed coupling between thyrotropin-releasing hormone (TRH) receptor and its cognate G(q/11) protein in HEK293 cells expressing high levels of these proteins. Under different solubilization conditions, the TRH receptor (TRH-R) was identified to form a putative pentameric complex composed of TRH-R homodimer and G(q/11) protein. The presumed association of TRH-R with G(q/11)α or Gß proteins in plasma membranes was verified by RNAi experiments. After 10- or 30-min hormone treatment, TRH-R signaling complexes gradually dissociated with a concomitant release of receptor homodimers. These observations support the model in which GPCRs can be coupled to trimeric G-proteins in preassembled signaling complexes, which might be dynamically regulated upon receptor activation. The precoupling of receptors with their cognate G-proteins can contribute to faster G-protein activation and subsequent signal transfer into the cell interior.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , Receptors, Thyrotropin-Releasing Hormone/chemistry , Cell Line , Cell Membrane/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , HEK293 Cells , Humans , Receptors, Thyrotropin-Releasing Hormone/genetics , Receptors, Thyrotropin-Releasing Hormone/metabolism , Signal Transduction , Thyrotropin-Releasing Hormone/genetics , Thyrotropin-Releasing Hormone/metabolism , Transfection
8.
Gen Comp Endocrinol ; 174(2): 80-8, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21827760

ABSTRACT

Molecular cloning of thyrotropin-releasing hormone receptors (TRHR) was performed in a teleost, the sockeye salmon (Oncorhynchus nerka). Four different TRHR cDNAs were cloned and named TRHR1, TRHR2a, TRHR2b and TRHR3 based on their similarity to known TRHR subtypes in vertebrates. Important residues for TRH binding were conserved in deduced amino acid sequences of the three TRHR subtypes except for the TRHR2b. Seven transmembrane domains were predicted for TRHR1, TRHR2a and TRHR3 proteins but only five for TRHR2b which appears to be truncated. In silico database analysis identified putative TRHR sequences including invertebrate TRHR and reptilian, avian and mammalian TRHR3. Phylogenetic analyses predicted the molecular evolution of TRHR in vertebrates: from the common ancestral TRHR (i.e. invertebrate TRHR), the TRHR2 subtype diverged first and then TRHR1 and TRHR3 diverged. Reverse transcription-polymerase chain reaction analyses revealed TRHR1 transcripts in the brain (hypothalamus), retina, pituitary gland and large intestine; TRHR2a in the brain (telencephalon and hypothalamus); and TRHR3 in the brain (olfactory bulbs) and retina.


Subject(s)
DNA, Complementary/genetics , Fish Proteins/genetics , Receptors, Thyrotropin-Releasing Hormone/genetics , Salmon/genetics , Amino Acid Sequence , Animals , Cloning, Molecular , Evolution, Molecular , Fish Proteins/chemistry , Fish Proteins/metabolism , Gene Expression/genetics , Molecular Sequence Data , Receptors, Thyrotropin-Releasing Hormone/chemistry , Receptors, Thyrotropin-Releasing Hormone/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, Protein
9.
Gen Comp Endocrinol ; 170(2): 374-80, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-20977909

ABSTRACT

Molecular cloning of thyrotropin-releasing hormone receptors (TRHR) was performed in a model teleost fish, medaka (Oryzias latipes). Four subtypes of TRHR were cloned and named them as TRHR1a, TRHR1b, TRHR2 and TRHR3 based on their similarity to known TRHR subtypes in vertebrates. TRHR1a, TRHR1b, TRHR2, and TRHR3 of medaka encode 416, 398, 451, and 386 amino acid residues, respectively. Comparison of cDNA sequences of medaka TRHR subtypes with respective genomic DNA sequences revealed gene structures: TRHR1a, TRHR1b and TRHR3genes consist of two exons while the TRH2 gene consists of five exons. Molecular phylogenetic analyses depicted the molecular evolution of TRHR in vertebrates: From the ancestral molecule, TRHR2 diverged first and then TRHR1 and TRHR3. Reverse transcription-polymerase chain reaction analyses revealed the sites of TRHR expression: Expression of TRHR1, TRHR1b and TRHR2 subtypes has been confirmed in the brain, pineal organ, retina and pituitary gland. In addition, TRHR1b is expressed in spleen, digestive tract and skin, and TRHR2 in testis, ovary and gill. TRHR3 is widely expressed in various tissues. These results indicate that in medaka, TRH might exert multiple functions mediated by different TRHR subtypes expressed in each tissue.


Subject(s)
Evolution, Molecular , Fish Proteins/genetics , Oryzias/genetics , Receptors, Thyrotropin-Releasing Hormone/genetics , Amino Acid Sequence , Animals , Cloning, Molecular , Female , Fish Proteins/chemistry , Fish Proteins/metabolism , Male , Molecular Sequence Data , Oryzias/metabolism , Phylogeny , Receptors, Thyrotropin-Releasing Hormone/chemistry , Receptors, Thyrotropin-Releasing Hormone/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Analysis, Protein
10.
Brain Res ; 1355: 70-85, 2010 Oct 08.
Article in English | MEDLINE | ID: mdl-20691166

ABSTRACT

We have reported a highly cooperative interaction between leptin and thyrotropin releasing hormone (TRH) in the hindbrain to generate thermogenic responses (Hermann et al., 2006) (Rogers et al., 2009). Identifying the locus in the hindbrain where leptin and TRH act synergistically to increase thermogenesis will be necessary before we can determine the mechanism(s) by which this interaction occurs. Here, we performed heat-induced epitope recovery techniques and in situ hybridization to determine if neurons or afferent fibers in the hindbrain possess both TRH type 1 receptor and long-form leptin receptor [TRHR1; LepRb, respectively]. LepRb receptors were highly expressed in the solitary nucleus [NST], dorsal motor nucleus of the vagus [DMN] and catecholaminergic neurons of the ventrolateral medulla [VLM]. All neurons that contained LepRb also contained TRHR1. Fibers in the NST and the raphe pallidus [RP] and obscurrus [RO] that possess LepRb receptors were phenotypically identified as glutamatergic type 2 fibers (vglut2). Fibers in the NST and RP that possess TRHR1 receptors were phenotypically identified as serotonergic [i.e., immunopositive for the serotonin transporter; SERT]. Co-localization of LepRb and TRHR1 was not observed on individual fibers in the hindbrain but these two fiber types co-mingle in these nuclei. These anatomical arrangements may provide a basis for the synergy between leptin and TRH to increase thermogenesis.


Subject(s)
Medulla Oblongata/metabolism , Neurons/metabolism , Receptors, Leptin/metabolism , Receptors, Thyrotropin-Releasing Hormone/metabolism , Rhombencephalon/metabolism , Animals , Female , Humans , Male , Medulla Oblongata/chemistry , Medulla Oblongata/cytology , Mice , Mice, Inbred C57BL , Mice, Obese , Neurons/chemistry , Neurons/cytology , Raphe Nuclei/physiology , Rats , Rats, Long-Evans , Receptors, Leptin/genetics , Receptors, Thyrotropin-Releasing Hormone/chemistry , Receptors, Thyrotropin-Releasing Hormone/genetics , Reticular Formation/cytology , Reticular Formation/metabolism , Rhombencephalon/chemistry , Rhombencephalon/cytology , Solitary Nucleus/cytology , Solitary Nucleus/metabolism , Vagus Nerve/cytology , Vagus Nerve/metabolism
11.
Mol Pharmacol ; 77(2): 288-97, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19906838

ABSTRACT

The thyrotropin-releasing hormone (TRH) receptor undergoes rapid and extensive agonist-dependent phosphorylation attributable to G protein-coupled receptor (GPCR) kinases (GRKs), particularly GRK2. Like many GPCRs, the TRH receptor is predicted to form an amphipathic helix, helix 8, between the NPXXY motif at the cytoplasmic end of the seventh transmembrane domain and palmitoylation sites at Cys335 and Cys337. Mutation of all six lysine and arginine residues between the NPXXY and residue 340 to glutamine (6Q receptor) did not prevent the receptor from stimulating inositol phosphate turnover but almost completely prevented receptor phosphorylation in response to TRH. Phosphorylation at all sites in the cytoplasmic tail was inhibited. The phosphorylation defect was not reversed by long incubation times or high TRH concentrations. As expected for a phosphorylation-defective receptor, the 6Q-TRH receptor did not recruit arrestin, undergo the typical arrestin-dependent increase in agonist affinity, or internalize well. Lys326, directly before phenylalanine in the common GPCR motif NPXXY(X)(5-6)F(R/K), was critical for phosphorylation. The 6Q-TRH receptor was not phosphorylated effectively in cells overexpressing GRK2 or in in vitro kinase assays containing purified GRK2. Phosphorylation of the 6Q receptor was partially restored by coexpression of a receptor with an intact helix 8 but without phosphorylation sites. Phosphorylation was inhibited but not completely prevented by alanine substitution for cysteine palmitoylation sites. Positively charged amino acids in the proximal tail of the beta2-adrenergic receptor were also important for GRK-dependent phosphorylation. The results indicate that positive residues in helix 8 of GPCRs are important for GRK-dependent phosphorylation.


Subject(s)
G-Protein-Coupled Receptor Kinases/chemistry , G-Protein-Coupled Receptor Kinases/physiology , Receptors, Thyrotropin-Releasing Hormone/chemistry , Receptors, Thyrotropin-Releasing Hormone/physiology , Amino Acid Sequence , Animals , Cell Line , Humans , Mice , Molecular Sequence Data , Phosphorylation/physiology , Protein Structure, Secondary/physiology
12.
Gen Comp Endocrinol ; 164(1): 40-50, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19435597

ABSTRACT

Thyrotropin-releasing hormone (TRH) is the first hypothalamic hypophysiotropic neuropeptide whose sequence has been chemically characterized. The primary structure of TRH (pGlu-His-Pro-NH(2)) has been fully conserved across the vertebrate phylum. TRH is generated from a large precursor protein that contains multiple repeats of the TRH progenitor tetrapeptide Gln-His-Pro-Gly. In all tetrapods, TRH-expressing neurons located in the hypothalamus project towards the external zone of the median eminence while in teleosts they directly innervate the pars distalis of the pituitary. In addition, in frogs and teleosts, a bundle of TRH-containing fibers terminate in the neurointermediate lobe of the pituitary gland. Although TRH was originally named for its ability to trigger the release of thyroid-stimulating hormone (TSH) in mammals, it later became apparent that it exerts multiple, species-dependent hypophysiotropic activities. Thus, in fish TRH stimulates growth hormone (GH) and prolactin (PRL) release but does not affect TSH secretion. In amphibians, TRH is a marginal stimulator of TSH release in adult frogs, not in tadpoles, and a major releasing factor for GH and PRL. In birds, TRH triggers TSH and GH secretion. In mammals, TRH stimulates TSH, GH and PRL release. In fish and amphibians, TRH is also a very potent stimulator of alpha-melanocyte-stimulating hormone release. Because the intermediate lobe of the pituitary of amphibians is composed by a single type of hormone-producing cells, the melanotrope cells, it is a suitable model in which to investigate the mechanism of action of TRH at the cellular and molecular level. The occurrence of large amounts of TRH in the frog skin and high concentrations of TRH in frog plasma suggests that, in amphibians, skin-derived TRH may exert hypophysiotropic functions.


Subject(s)
Thyrotropin-Releasing Hormone/physiology , Animals , Growth Hormone/metabolism , Humans , Models, Biological , Pituitary Gland/metabolism , Prolactin/metabolism , Receptors, Thyrotropin-Releasing Hormone/chemistry , Receptors, Thyrotropin-Releasing Hormone/genetics , Receptors, Thyrotropin-Releasing Hormone/metabolism , Thyrotropin/metabolism , Thyrotropin-Releasing Hormone/metabolism
13.
J Biomol Screen ; 13(9): 888-98, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18812574

ABSTRACT

The bioluminescence resonance energy transfer (BRET) technique has become extremely popular for studying protein-protein interactions in living cells and real time. Of particular interest is the ability to monitor interactions between G protein-coupled receptors, such as the thyrotropin-releasing hormone receptor (TRHR), and proteins critical for regulating their function, such as beta-arrestin. Using TRHR/beta-arrestin interactions, we have demonstrated improvements to all 3 generations of BRET (BRET(1), BRET(2), and eBRET) by using the novel forms of luciferase, Rluc2 and Rluc8, developed by the Gambhir laboratory. Furthermore, for the 1st time it was possible to use the BRET2 system to detect ligand-induced G protein-coupled receptor/beta-arrestin interactions over prolonged periods (on the scale of hours rather than seconds) with a very stable signal. As demonstrated by our Z'-factor data, these luciferases increase the sensitivity of BRET to such an extent that they substantially increase the potential applicability of this technology for effective drug discovery high-throughput screening.


Subject(s)
Energy Transfer , Receptors, G-Protein-Coupled/chemistry , Cell Line , Drug Design , Drug Discovery , Humans , Inositol Phosphates/chemistry , Kinetics , Luciferases/chemistry , Luminescent Proteins/chemistry , Receptors, Thyrotropin-Releasing Hormone/chemistry , Recombinant Fusion Proteins/chemistry , Time Factors
14.
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
15.
Mol Pharmacol ; 74(1): 195-202, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18413662

ABSTRACT

Arrestin binding to agonist-occupied phosphorylated G protein-coupled receptors typically increases the affinity of agonist binding, increases resistance of receptor-bound agonist to removal with high acid/salt buffer, and leads to receptor desensitization and internalization. We tested whether thyrotropin-releasing hormone (TRH) receptors lacking phosphosites in the C-terminal tail could form stable and functional complexes with arrestin. Fibroblasts from mice lacking arrestins 2 and 3 were used to distinguish between arrestin-dependent and -independent effects. Arrestin did not promote internalization or desensitization of a receptor that had key Ser/Thr phosphosites mutated to Ala (4Ala receptor). Nevertheless, arrestin greatly increased acid/salt resistance and the affinity of 4Ala receptor for TRH. Truncation of 4Ala receptor just distal to the key phosphosites (4AlaStop receptor) abolished arrestin-dependent acid/salt resistance but not the effect of arrestin on agonist affinity. Arrestin formed stable complexes with activated wild-type and 4Ala receptors but not with 4AlaStop receptor, as measured by translocation of arrestin-green fluorescent protein to the plasma membrane or chemical cross-linking. An arrestin mutant that does not interact with clathrin and AP2 did not internalize receptor but still promoted high affinity TRH binding, acid/salt resistance, and desensitization. A sterically restricted arrestin mutant did not cause receptor internalization or desensitization but did promote acid/salt resistance and high agonist affinity. The results demonstrate that arrestin binds to proximal or distal phosphosites in the receptor tail. Arrestin binding at either site causes increased agonist affinity and acid/salt resistance, but only the proximal phosphosites evoke the necessary conformational changes in arrestin for receptor desensitization and internalization.


Subject(s)
Arrestin/metabolism , Receptors, Thyrotropin-Releasing Hormone/metabolism , Animals , Arrestin/chemistry , Arrestin/genetics , Cell Line , Cells, Cultured , Embryo, Mammalian , Fibroblasts/metabolism , Green Fluorescent Proteins/metabolism , Hemagglutinins/metabolism , Humans , Inositol Phosphates/biosynthesis , Kidney/cytology , Ligands , Mice , Models, Biological , Mutation , Phosphorylation , Protein Structure, Tertiary , Receptors, Thyrotropin-Releasing Hormone/chemistry , Transfection
16.
Proteins ; 71(2): 783-94, 2008 May 01.
Article in English | MEDLINE | ID: mdl-17979196

ABSTRACT

Multiple computational methods have been employed in a comparative study of thyrotropin-releasing hormone receptors 1 and 2 (TRH-R1 and TRH-R2) to explore the structural bases for the different functional properties of these G protein-coupled receptors. Three-dimensional models of both murine TRH receptors have been built and optimized by means of homology modeling based on the crystal structure of bovine rhodopsin, molecular dynamics simulations, and energy minimizations in a membrane-aqueous environment. The comparison between the two models showed a correlation between the higher flexibility and higher basal activity of TRH-R2 versus the lesser flexibility and lower basal activity of TRH-R1 and supported the involvement of the highly conserved W6.48 in the signaling process. A correlation between the level of basal activity and conformational changes of TM5 was detected also. Comparison between models of the wild type receptors and their W6.48A mutants, which have reversed basal activities compared with their respective wild types, further supported these correlations. A flexible molecular docking procedure revealed that TRH establishes a direct interaction with W6.48 in TRH-R2 but not in TRH-R1. We designed and performed new mutagenesis experiments that strongly supported these observations.


Subject(s)
Receptors, Thyrotropin-Releasing Hormone/chemistry , Receptors, Thyrotropin-Releasing Hormone/physiology , Amino Acid Sequence , Animals , Binding Sites , Cattle , Computer Simulation , Crystallography, X-Ray , Mice , Models, Molecular , Molecular Sequence Data , Monte Carlo Method , Protein Binding , Protein Conformation , Receptors, Thyrotropin-Releasing Hormone/genetics , Rhodopsin/chemistry , Sequence Alignment , Thyrotropin-Releasing Hormone/metabolism
17.
Proc Natl Acad Sci U S A ; 104(46): 18303-8, 2007 Nov 13.
Article in English | MEDLINE | ID: mdl-17989235

ABSTRACT

The G protein-coupled thyrotropin (TSH)-releasing hormone (TRH) receptor forms homodimers. Regulated receptor dimerization increases TRH-induced receptor endocytosis. These studies test whether dimerization increases receptor phosphorylation, which could potentiate internalization. Phosphorylation at residues 355-365, which is critical for internalization, was measured with a highly selective phospho-site-specific antibody. Two strategies were used to drive receptor dimerization. Dimerization of a TRH receptor-FK506-binding protein (FKBP) fusion protein was stimulated by a dimeric FKBP ligand. The chemical dimerizer caused a large increase in TRH-dependent phosphorylation within 1 min, whereas a monomeric FKBP ligand had no effect. The dimerizer did not alter phoshorylation of receptors lacking the FKBP domain. Dimerization of receptors containing an N-terminal HA epitope also was induced with anti-HA antibody. Anti-HA IgG strongly increased TRH-induced phosphorylation, whereas monomeric Fab fragments had no effect. Anti-HA antibody did not alter phosphorylation in receptors lacking an HA tag. Furthermore, two phosphorylation-defective TRH receptors functionally complemented one another and permitted phosphorylation. Receptors with a D71A mutation in the second transmembrane domain do not signal, whereas receptors with four Ala mutations in the 355-365 region signal normally but lack phosphorylation sites. When D71A- and 4Ala-TRH receptors were expressed alone, neither underwent TRH-dependent phosphorylation. When they were expressed together, D71A receptor was phosphorylated by G protein-coupled receptor kinases in response to TRH. These results suggest that the TRH receptor is phosphorylated preferentially when it is in dimers or when preexisting receptor dimers are driven into microaggregates. Increased receptor phosphorylation may amplify desensitization.


Subject(s)
Receptors, Thyrotropin-Releasing Hormone/metabolism , Dimerization , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Phosphorylation , Receptors, Thyrotropin-Releasing Hormone/chemistry
18.
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
19.
Pharmacol Ther ; 113(2): 410-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17123625

ABSTRACT

Thyrotropin-releasing hormone (TRH), a tripeptide, exerts its biological effects through stimulation of cell-surface receptors, TRH-R, belonging to the superfamily of G protein-coupled receptors (GPCR). Because of the intermediate size of TRH, it is smaller than polypeptide ligands that interact at GPCR ectodomains and larger than biogenic amines, which interact within GPCR transmembrane domains (TMD), the TRH/TRH-R complex probably shares properties of these 2 extremes, representing a unique system to study GPCR/ligand interactions. In this review, we summarize the current knowledge of the structure-activity relationships in the TRH/TRH-R system. Based on experimental data and the structural information acquired from computer simulations, we formulate a working hypothesis to describe the molecular events underlying the processes of TRH binding and TRH-R activation. This hypothesis represents a starting point for understanding the biology of the TRH/TRH-R system on a molecular level and provides a basis for potential design of new potent and selective modulators of TRH-R's activity.


Subject(s)
Receptors, Thyrotropin-Releasing Hormone/metabolism , Thyrotropin-Releasing Hormone/metabolism , Animals , Binding Sites , Models, Molecular , Protein Conformation , Receptors, Thyrotropin-Releasing Hormone/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...