59 results on '"Receptors, Thyrotropin-Releasing Hormone chemistry"'
Search Results
2. Characterization of a novel thyrotropin-releasing hormone receptor, TRHR3, in chickens.
- Author
-
Li X, Li Z, Deng Y, Zhang J, Li J, and Wang Y
- Subjects
- 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, Amino Acid Sequence, Chickens genetics, Receptors, Thyrotropin-Releasing Hormone genetics
- 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., (Copyright © 2019. Published by Elsevier Inc.)
- Published
- 2020
- Full Text
- View/download PDF
3. TRH receptor mobility in the plasma membrane is strongly affected by agonist binding and by interaction with some cognate signaling proteins.
- Author
-
Moravcova R, Melkes B, and Novotny J
- Subjects
- 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, GTP-Binding Protein alpha Subunits, Gi-Go chemistry, Receptors, Thyrotropin-Releasing Hormone chemistry, Signal Transduction drug effects, beta-Arrestins chemistry
- 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 G
q/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/11 α, Gβ, β-arrestin2 and phospholipase Cβ1, but not of Gi α1, β-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 Gi α1 and β-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.- Published
- 2018
- Full Text
- View/download PDF
4. Synthesis and biology of ring-modified l-Histidine containing thyrotropin-releasing hormone (TRH) analogues.
- Author
-
Meena CL, Thakur A, Nandekar PP, Sharma SS, Sangamwar AT, and Jain R
- Subjects
- HEK293 Cells, Histidine chemistry, Humans, Models, Molecular, Molecular Conformation, Receptors, Thyrotropin-Releasing Hormone chemistry, Receptors, Thyrotropin-Releasing Hormone metabolism, Thyrotropin-Releasing Hormone chemistry, Histidine metabolism, Thyrotropin-Releasing Hormone analogs & derivatives, Thyrotropin-Releasing Hormone metabolism
- 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., (Copyright © 2016 Elsevier Masson SAS. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
5. Synthesis of CNS active thyrotropin-releasing hormone (TRH)-like peptides: Biological evaluation and effect on cognitive impairment induced by cerebral ischemia in mice.
- Author
-
Meena CL, Thakur A, Nandekar PP, Sangamwar AT, Sharma SS, and Jain R
- Subjects
- Animals, Brain Ischemia, Mice, Molecular Structure, Rats, Cognition Disorders genetics, Peptides metabolism, Receptors, Thyrotropin-Releasing Hormone chemistry, Thyrotropin-Releasing Hormone chemical synthesis
- 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., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
6. TRH-receptor mobility and function in intact and cholesterol-depleted plasma membrane of HEK293 cells stably expressing TRH-R-eGFP.
- Author
-
Brejchová J, Sýkora J, Ostašov P, Merta L, Roubalová L, Janáček J, Hof M, and Svoboda P
- Subjects
- 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, Cell Membrane metabolism, Cholesterol metabolism, Green Fluorescent Proteins metabolism, Receptors, Thyrotropin-Releasing Hormone metabolism
- 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., (Copyright © 2014 Elsevier B.V. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
7. Identification of a preassembled TRH receptor-G(q/11) protein complex in HEK293 cells.
- Author
-
Drastichova Z and Novotny J
- Subjects
- 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, GTP-Binding Protein alpha Subunits, Gq-G11 chemistry, Receptors, Thyrotropin-Releasing Hormone chemistry
- 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 Gβ 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.
- Published
- 2012
- Full Text
- View/download PDF
8. Molecular cloning, molecular evolution and gene expression of cDNAs encoding thyrotropin-releasing hormone receptor subtypes in a teleost, the sockeye salmon (Oncorhynchus nerka).
- Author
-
Saito Y, Mekuchi M, Kobayashi N, Kimura M, Aoki Y, Masuda T, Azuma T, Fukami M, Iigo M, and Yanagisawa T
- Subjects
- 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, DNA, Complementary genetics, Fish Proteins genetics, Receptors, Thyrotropin-Releasing Hormone genetics, Salmon genetics
- 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., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
9. Molecular cloning, gene structure, molecular evolution and expression analyses of thyrotropin-releasing hormone receptors from medaka (Oryzias latipes).
- Author
-
Mekuchi M, Saito Y, Aoki Y, Masuda T, Iigo M, and Yanagisawa T
- Subjects
- 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, Evolution, Molecular, Fish Proteins genetics, Oryzias genetics, Receptors, Thyrotropin-Releasing Hormone genetics
- 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., (Copyright © 2010 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
10. Co-localization of TRHR1 and LepRb receptors on neurons in the hindbrain of the rat.
- Author
-
Barnes MJ, Rogers RC, Van Meter MJ, and Hermann GE
- Subjects
- 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, Medulla Oblongata metabolism, Neurons metabolism, Receptors, Leptin metabolism, Receptors, Thyrotropin-Releasing Hormone metabolism, Rhombencephalon metabolism
- 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., (Copyright © 2010 Elsevier B.V. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
11. Role of helix 8 of the thyrotropin-releasing hormone receptor in phosphorylation by G protein-coupled receptor kinase.
- Author
-
Gehret AU, Jones BW, Tran PN, Cook LB, Greuber EK, and Hinkle PM
- Subjects
- Amino Acid Sequence, Animals, Cell Line, Humans, Mice, Molecular Sequence Data, Phosphorylation physiology, Protein Structure, Secondary physiology, G-Protein-Coupled Receptor Kinases chemistry, G-Protein-Coupled Receptor Kinases physiology, Receptors, Thyrotropin-Releasing Hormone chemistry, Receptors, Thyrotropin-Releasing Hormone physiology
- 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.
- Published
- 2010
- Full Text
- View/download PDF
12. TRH acts as a multifunctional hypophysiotropic factor in vertebrates.
- Author
-
Galas L, Raoult E, Tonon MC, Okada R, Jenks BG, Castaño JP, Kikuyama S, Malagon M, Roubos EW, and Vaudry H
- Subjects
- 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, Thyrotropin-Releasing Hormone physiology
- 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.
- Published
- 2009
- Full Text
- View/download PDF
13. Selectivity-based QSAR approach for screening and evaluation of TRH analogs for TRH-R1 and TRH-R2 receptors subtypes.
- Author
-
Kadam RU, Chavan AG, Monga V, Kaur N, Jain R, and Roy N
- Subjects
- Models, Molecular, Receptors, Thyrotropin-Releasing Hormone chemistry, Reproducibility of Results, Drug Design, Drug Evaluation, Quantitative Structure-Activity Relationship, Receptors, Thyrotropin-Releasing Hormone agonists, Thyrotropin-Releasing Hormone analogs & derivatives
- 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.
- Published
- 2008
- Full Text
- View/download PDF
14. Demonstration of improvements to the bioluminescence resonance energy transfer (BRET) technology for the monitoring of G protein-coupled receptors in live cells.
- Author
-
Kocan M, See HB, Seeber RM, Eidne KA, and Pfleger KD
- Subjects
- 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, Energy Transfer, Receptors, G-Protein-Coupled chemistry
- 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.
- Published
- 2008
- Full Text
- View/download PDF
15. Arrestin binds to different phosphorylated regions of the thyrotropin-releasing hormone receptor with distinct functional consequences.
- Author
-
Jones BW and Hinkle PM
- Subjects
- 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, Arrestin metabolism, Receptors, Thyrotropin-Releasing Hormone metabolism
- 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.
- Published
- 2008
- Full Text
- View/download PDF
16. Understanding the structural and functional differences between mouse thyrotropin-releasing hormone receptors 1 and 2.
- Author
-
Deflorian F, Engel S, Colson AO, Raaka BM, Gershengorn MC, and Costanzi S
- Subjects
- 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, Receptors, Thyrotropin-Releasing Hormone chemistry, Receptors, Thyrotropin-Releasing Hormone physiology
- 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.
- Published
- 2008
- Full Text
- View/download PDF
17. Dimerization of the thyrotropin-releasing hormone receptor potentiates hormone-dependent receptor phosphorylation.
- Author
-
Song GJ, Jones BW, and Hinkle PM
- Subjects
- Dimerization, Enzyme-Linked Immunosorbent Assay, Fluorescent Antibody Technique, Phosphorylation, Receptors, Thyrotropin-Releasing Hormone chemistry, Receptors, Thyrotropin-Releasing Hormone metabolism
- 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.
- Published
- 2007
- Full Text
- View/download PDF
18. Thyrotropin-releasing hormone and its receptors--a hypothesis for binding and receptor activation.
- Author
-
Engel S and Gershengorn MC
- Subjects
- Animals, Binding Sites, Models, Molecular, Protein Conformation, Receptors, Thyrotropin-Releasing Hormone chemistry, Receptors, Thyrotropin-Releasing Hormone metabolism, Thyrotropin-Releasing Hormone metabolism
- 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.
- Published
- 2007
- Full Text
- View/download PDF
19. Modifications of the pyroglutamic acid and histidine residues in thyrotropin-releasing hormone (TRH) yield analogs with selectivity for TRH receptor type 2 over type 1.
- Author
-
Kaur N, Monga V, Lu X, Gershengorn MC, and Jain R
- Subjects
- Molecular Conformation, Protein Binding, Receptors, Thyrotropin-Releasing Hormone chemistry, Stereoisomerism, Structure-Activity Relationship, Thyrotropin-Releasing Hormone analogs & derivatives, Histidine chemistry, Pyrrolidonecarboxylic Acid chemistry, Receptors, Thyrotropin-Releasing Hormone agonists, Thyrotropin-Releasing Hormone chemical synthesis, Thyrotropin-Releasing Hormone pharmacology
- 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.
- Published
- 2007
- Full Text
- View/download PDF
20. [Congenital TSH * PRL combined deficiency (TRH receptor gene mutation)].
- Author
-
Hashimoto K and Yamada M
- Subjects
- Animals, Diagnosis, Differential, Humans, Hypopituitarism diagnosis, Hypopituitarism physiopathology, Hypopituitarism therapy, Prognosis, Receptors, Thyrotropin-Releasing Hormone chemistry, Receptors, Thyrotropin-Releasing Hormone physiology, Thyroxine administration & dosage, Hypopituitarism etiology, Mutation, Prolactin deficiency, Receptors, Thyrotropin-Releasing Hormone genetics, Thyrotropin deficiency
- Published
- 2006
21. Thyrotropin-releasing hormone analogs.
- Author
-
Colson AO and Gershengorn MC
- Subjects
- Animals, Central Nervous System drug effects, Humans, Receptors, Thyrotropin-Releasing Hormone chemistry, Thyrotropin-Releasing Hormone pharmacology, Thyrotropin-Releasing Hormone analogs & derivatives
- Abstract
Thyrotropin releasing hormone (TRH: pyroglutamic acid-histidine-prolineamide) regulates the activity of cells in the anterior pituitary and within the central and peripheral nervous systems. TRH, which has been the subject of much research over the past three decades, exerts its effects by acting through class A G-protein coupled receptors. The recent discovery of a second receptor subtype has generated an interest in the discovery of receptor subtype-selective TRH analogs. In this review, we describe advances in the development of TRH analogs and in the understanding of their mechanism of interaction with TRH receptors. We also describe the recent breakthrough in the identification of analogs that bind selectively at TRH-R2.
- Published
- 2006
- Full Text
- View/download PDF
22. Beta-arrestin mediates desensitization and internalization but does not affect dephosphorylation of the thyrotropin-releasing hormone receptor.
- Author
-
Jones BW and Hinkle PM
- Subjects
- Alkaline Phosphatase metabolism, Animals, Arrestins chemistry, Arrestins metabolism, CHO Cells, COS Cells, Calcium Channels metabolism, Cell Line, Cell Membrane metabolism, Chlorocebus aethiops, Cricetinae, DNA, Complementary metabolism, Dose-Response Relationship, Drug, Endocytosis, Fibroblasts metabolism, G-Protein-Coupled Receptor Kinase 2, GTP-Binding Protein alpha Subunits, Gq-G11 metabolism, GTP-Binding Proteins metabolism, Glycosylation, Green Fluorescent Proteins metabolism, Immunoblotting, Immunoglobulin G chemistry, Immunoprecipitation, Inositol 1,4,5-Trisphosphate metabolism, Inositol 1,4,5-Trisphosphate Receptors, Inositol Phosphates chemistry, Inositol Phosphates metabolism, Kinetics, Mice, Mice, Knockout, Mutation, Phosphates chemistry, Phosphorylation, Plasmids metabolism, Protein Binding, Protein Kinase C antagonists & inhibitors, Protein Kinase C metabolism, Protein Structure, Tertiary, Protein Transport, Receptors, Cytoplasmic and Nuclear metabolism, Receptors, Thyrotropin-Releasing Hormone chemistry, Sucrose chemistry, Sucrose pharmacology, Time Factors, Transfection, beta-Adrenergic Receptor Kinases metabolism, beta-Arrestin 1, beta-Arrestin 2, beta-Arrestins, Arrestins physiology, Receptors, Thyrotropin-Releasing Hormone physiology
- Abstract
The G protein-coupled thyrotropin-releasing hormone (TRH) receptor is phosphorylated and binds to beta-arrestin after agonist exposure. To define the importance of receptor phosphorylation and beta-arrestin binding in desensitization, and to determine whether beta-arrestin binding and receptor endocytosis are required for receptor dephosphorylation, we expressed TRH receptors in fibroblasts from mice lacking beta-arrestin-1 and/or beta-arrestin-2. Apparent affinity for [(3)H]MeTRH was increased 8-fold in cells expressing beta-arrestins, including a beta-arrestin mutant that did not permit receptor internalization. TRH caused extensive receptor endocytosis in the presence of beta-arrestins, but receptors remained primarily on the plasma membrane without beta-arrestin. beta-Arrestins strongly inhibited inositol 1,4,5-trisphosphate production within 10 s. At 30 min, endogenous beta-arrestins reduced TRH-stimulated inositol phosphate production by 48% (beta-arrestin-1), 71% (beta-arrestin-2), and 84% (beta-arrestins-1 and -2). In contrast, receptor phosphorylation, detected by the mobility shift of deglycosylated receptor, was unaffected by beta-arrestins. Receptors were fully phosphorylated within 15 s of TRH addition. Receptor dephosphorylation was identical with or without beta-arrestins and almost complete 20 min after TRH withdrawal. Blocking endocytosis with hypertonic sucrose did not alter the rate of receptor phosphorylation or dephosphorylation. Expressing receptors in cells lacking Galpha(q) and Galpha(11) or inhibiting protein kinase C pharmacologically did not prevent receptor phosphorylation or dephosphorylation. Overexpression of dominant negative G protein-coupled receptor kinase-2 (GRK2), however, retarded receptor phosphorylation. Receptor activation caused translocation of endogenous GRK2 to the plasma membrane. The results show conclusively that receptor dephosphorylation can take place on the plasma membrane and that beta-arrestin binding is critical for desensitization and internalization.
- Published
- 2005
- Full Text
- View/download PDF
23. Sequential phosphorylation mediates receptor- and kinase-induced inhibition of TREK-1 background potassium channels.
- Author
-
Murbartián J, Lei Q, Sando JJ, and Bayliss DA
- Subjects
- Alanine chemistry, Animals, Aspartic Acid chemistry, Binding Sites, Blotting, Western, Cell Line, Cell Membrane metabolism, Cloning, Molecular, Colforsin pharmacology, Cyclic AMP analogs & derivatives, Cyclic AMP pharmacology, Cyclic AMP-Dependent Protein Kinases antagonists & inhibitors, Humans, Indoles pharmacology, Maleimides pharmacology, Mice, Models, Biological, Mutagenesis, Site-Directed, Mutation, Orexin Receptors, Phorbol 12,13-Dibutyrate pharmacology, Phosphorylation, Potassium Channels chemistry, Potassium Channels, Tandem Pore Domain metabolism, Protein Binding, Protein Kinase C antagonists & inhibitors, Protein Kinase C metabolism, Protein Structure, Tertiary, Receptors, G-Protein-Coupled, Receptors, Neuropeptide chemistry, Receptors, Thyrotropin-Releasing Hormone chemistry, Recombinant Proteins chemistry, Serine chemistry, Time Factors, Transfection, Potassium chemistry, Potassium Channels, Tandem Pore Domain chemistry
- Abstract
Background potassium channels determine membrane potential and input resistance and serve as prominent effectors for modulatory regulation of cellular excitability. TREK-1 is a two-pore domain background K+ channel (KCNK2, K2P2.1) that is sensitive to a variety of physicochemical and humoral factors. In this work, we used a recombinant expression system to show that activation of G alpha(q)-coupled receptors leads to inhibition of TREK-1 channels via protein kinase C (PKC), and we identified a critical phosphorylation site in a key regulatory domain that mediates inhibition of the channel. In HEK 293 cells co-expressing TREK-1 and either the thyrotropin-releasing hormone receptor (TRHR1) or the Orexin receptor (Orx1R), agonist stimulation induced robust channel inhibition that was suppressed by a bisindolylmaleimide PKC inhibitor but not by a protein kinase A blocker ((R(p))-cAMP-S). Channel inhibition by agonists or by direct activators of PKC (phorbol dibutyrate) and PKA (forskolin) was disrupted not only by alanine or aspartate mutations at an identified PKA site (Ser-333) in the C terminus, but also at a more proximal regulatory site in the cytoplasmic C terminus (Ser-300); S333A and S300A mutations enhanced basal TREK-1 current, whereas S333D and S300D substitutions mimicked phosphorylation and strongly diminished currents. When studied in combination, TREK-1 current density was enhanced in S300A/S333D but reduced in S300D/S333A mutant channels. Channel mutants were expressed and appropriately targeted to cell membranes. Together, these data support a sequential phosphorylation model in which receptor-induced kinase activation drives modification at Ser-333 that enables subsequent phosphorylation at Ser-300 to inhibit TREK-1 channel activity.
- Published
- 2005
- Full Text
- View/download PDF
24. Dominant portion of thyrotropin-releasing hormone receptor is excluded from lipid domains. Detergent-resistant and detergent-sensitive pools of TRH receptor and Gqalpha/G11alpha protein.
- Author
-
Rudajev V, Novotny J, Hejnova L, Milligan G, and Svoboda P
- Subjects
- Caveolin 1, Caveolins chemistry, Cell Culture Techniques, Centrifugation, Density Gradient, Detergents, GTP-Binding Protein alpha Subunits, Gq-G11 analysis, Humans, Immunoblotting, Octoxynol, Radioligand Assay, Receptors, Thyrotropin-Releasing Hormone analysis, Solubility, Spectrometry, Fluorescence, Cell Membrane chemistry, GTP-Binding Protein alpha Subunits, Gq-G11 chemistry, Membrane Microdomains chemistry, Receptors, Thyrotropin-Releasing Hormone chemistry
- Abstract
Some G protein-coupled receptors might be spacially targetted to discrete domains within the plasma membrane. Here we assessed the localization in membrane domains of the epitope-tagged, fluorescent version of thyrotropin-releasing hormone receptor (VSV-TRH-R-GFP) expressed in HEK293 cells. Our comparison of three different methods of cell fractionation (detergent extraction, alkaline treatment/sonication and mechanical homogenization) indicated that the dominant portion of plasma membrane pool of the receptor was totally solubilized by Triton X-100 and its distribution was similar to that of transmembrane plasma membrane proteins (glycosylated and non-glycosylated forms of CD147, MHCI, CD29, CD44, transmembrane form of CD58, Tapa1 and Na,K-ATPase). As expected, caveolin and GPI-bound proteins CD55, CD59 and GPI-bound form of CD58 were preferentially localized in detergent-resistant membrane domains (DRMs). Trimeric G proteins G(q)alpha/G(11)alpha, G(i)alpha1/G(i)alpha2, G(s)alphaL/G(s)alphaS and Gbeta were distributed almost equally between detergent-resistant and detergent-solubilized pools. In contrast, VSV-TRH-R-GFP, Galpha, Gbeta and caveolin were localized massively only in low-density membrane fragments of plasma membranes, which were generated by alkaline treatment/sonication or by mechanical homogenization of cells. These data indicate that VSV-TRH-R-GFP as well as other transmembrane markers of plasma membranes are excluded from TX-100-resistant, caveolin-enriched membrane domains. Trimeric G protein G(q)alpha/G(11)alpha occurs in both DRMs and in the bulk of plasma membranes, which is totally solubilized by TX-100.
- Published
- 2005
- Full Text
- View/download PDF
25. Agonist-induced conformational changes in thyrotropin-releasing hormone receptor type I: disulfide cross-linking and molecular modeling approaches.
- Author
-
Huang W, Osman R, and Gershengorn MC
- Subjects
- 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, Cross-Linking Reagents chemistry, Disulfides chemistry, Models, Molecular, Receptors, Thyrotropin-Releasing Hormone agonists, Receptors, Thyrotropin-Releasing Hormone chemistry, Thyrotropin-Releasing Hormone chemistry
- 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.
- Published
- 2005
- Full Text
- View/download PDF
26. The computer modelling of human TRH receptor, TRH and TRH-like peptides.
- Author
-
Bílek R and Stárka L
- Subjects
- Amino Acid Sequence genetics, Humans, Molecular Sequence Data, Peptide Fragments genetics, Protein Structure, Secondary genetics, Receptors, Thyrotropin-Releasing Hormone genetics, Thyrotropin-Releasing Hormone genetics, Computer Simulation, Models, Molecular, Peptide Fragments chemistry, Receptors, Thyrotropin-Releasing Hormone chemistry, Thyrotropin-Releasing Hormone chemistry
- Abstract
The aim of this work was to verify the possibility of interactions between the human TRH receptor (an integral membrane protein which belongs to family 1 of G-protein coupled receptors) and TRH-like peptides presented in the prostate gland. These peptides are characterized by substitution of basic amino acid histidine (related to authentic TRH) for neutral or acidic amino acid, such as glutamic acid, phenylalanine, glutamine or tyrosine. The physiological function of TRH-like peptides in peripheral tissues is not precisely known. However, according to our recent experiments, we assume the existence of a local hormonal network formed by TRH-like peptides and TSH in the prostate gland. The network can be associated with circulating thyroid and steroid hormones, and may represent a new regulatory mechanism influencing the proliferative ability of prostatic tissue. A similar network of authentic TRH and TSH was already found in the gastrointestinal tract. The experimentally determined 3D-structures of human TRH receptor (hTRHr) and TRH-like peptides are not available. From this point of view we used de novo modeling procedures of G-protein coupled receptors on an automated protein modeling server used at the Glaxo Wellcome Experimental Research (Geneva, Switzerland). 3D-structures of TRH-like peptides were determined with a computer program CORINA (written by the team of J. Gasteiger, Computer-Chemie-Centrum and Institute for Organic Chemistry, University of Erlangen-Nurenberg, Germany). The generated PDB files with 3D-coordinates were visualized with Swiss-Pdb Viewer Release 3.51 (Glaxo Wellcome). From recent results it is evident that polar amino acids belonging to the extracellular terminus of hTRHr transmembrane regions can participate in interactions between TRH and hTRHr. There is no direct evidence that TRH-like peptides interact with the presented hTRHr model. On the contrary, with respect to the similar 3D-shape and the identity of terminal amino acids, it appears that these interactions are highly probable as well as the nearly 100 % cross-reactions between TRH or TRH-like peptides and antibody specific against authentic TRH. Closed terminal amino acids (pyroglutamic acid and proline-amide) of TRH or TRH-like peptides are important for these interactions. Desamido-TRH or glutamyl metabolites will be repelled by the negative potential of ASP195 (E: D93) and GLU298 (G: E137).
- Published
- 2005
27. A model of inverse agonist action at thyrotropin-releasing hormone receptor type 1: role of a conserved tryptophan in helix 6.
- Author
-
Lu X, Huang W, Worthington S, Drabik P, Osman R, and Gershengorn MC
- Subjects
- Benzodiazepines pharmacology, Cells, Cultured, Humans, Models, Molecular, Protein Structure, Secondary, Receptors, Thyrotropin-Releasing Hormone chemistry, Receptors, Thyrotropin-Releasing Hormone drug effects, Signal Transduction drug effects, Thyrotropin-Releasing Hormone metabolism, Transfection, Midazolam pharmacology, Receptors, Thyrotropin-Releasing Hormone metabolism, Tryptophan metabolism
- Abstract
A binding pocket for thyrotropin-releasing hormone (TRH) within the transmembrane helices of the TRH receptor type 1 (TRH-R1) has been identified based on experimental evidence and computer simulations. To determine the binding site for a competitive inverse agonist, midazolam, three of the four residues that directly contact TRH and other residues that restrain TRH-R1 in an inactive conformation were screened by mutagenesis and binding assays. We found that two residues that directly contact TRH, Asn-110 in transmembrane helix 3 (3.37) and Arg-306 in transmembrane helix 7 (7.39), were important for midazolam binding but another, Tyr-282 in transmembrane helix 6 (6.51), was not. A highly conserved residue, Trp-279 in transmembrane helix 6 (6.48), which was reported to be critical in stabilizing TRH-R1 in an inactive state but not for TRH binding, was critical for midazolam binding. We used our previous model of the unoccupied TRH-R1 to generate a model of the TRH-R1/midazolam complex. The experimental results and the molecular model of the complex suggest that midazolam binds to TRH-R1 within a transmembrane helical pocket that partially overlaps the TRH binding pocket. This result is consistent with the competitive antagonism of midazolam binding. We suggest that the mechanism of inverse agonism effected by midazolam involves its direct interaction with Trp-279, which contributes to the stabilization of the inactive conformation of TRH-R1.
- Published
- 2004
- Full Text
- View/download PDF
28. Receptors for hypothalamic releasing hormones TRH and GnRH: oligomerization and interactions with intracellular proteins.
- Author
-
Pfleger KD, Kroeger KM, and Eidne KA
- Subjects
- Animals, Humans, Protein Binding, Protein Structure, Quaternary, Receptors, LHRH genetics, Receptors, Thyrotropin-Releasing Hormone genetics, Intracellular Space metabolism, Proteins metabolism, Receptors, LHRH chemistry, Receptors, LHRH metabolism, Receptors, Thyrotropin-Releasing Hormone chemistry, Receptors, Thyrotropin-Releasing Hormone metabolism
- Abstract
Studies of TRH and GnRH receptors have revealed much information about the roles of G-proteins and beta-arrestins, as well as receptor residues important for signaling, desensitization and internalization. However, the proteins involved are only just beginning to be identified and characterized. Additional complexity now exists with the observation that these receptors form oligomers in live cells. Indeed, hetero-oligomerization of TRH receptor subtypes 1 and 2 potentially alters interactions with intracellular regulatory proteins. Knowledge of proteins that interact with TRH or GnRH receptors will increase our understanding of receptor function and provide potential drug targets for a range of receptor-associated conditions.
- Published
- 2004
- Full Text
- View/download PDF
29. Thyrotropin-releasing hormone receptors -- similarities and differences.
- Author
-
Sun Y, Lu X, and Gershengorn MC
- Subjects
- Animals, Brain anatomy & histology, Brain metabolism, GTP-Binding Proteins metabolism, Humans, Protein Binding, Protein Conformation, Protein Isoforms chemistry, Protein Isoforms genetics, Receptors, Thyrotropin-Releasing Hormone chemistry, Receptors, Thyrotropin-Releasing Hormone genetics, Thyrotropin-Releasing Hormone analogs & derivatives, Tissue Distribution, Protein Isoforms metabolism, Receptors, Thyrotropin-Releasing Hormone metabolism, Signal Transduction physiology, Thyrotropin-Releasing Hormone metabolism
- Abstract
Thyrotropin-releasing hormone (TRH) initiates its effects by interacting with cell-surface membrane receptors. Two G protein-coupled receptors for TRH, TRH receptor type 1 (TRH-R1) and TRH receptor type 2 (TRH-R2), have been cloned from mammals. In this review, we compare TRH-R1 and TRH-R2 with regard to their tIssue distribution, binding affinities for TRH and TRH analogs, basal and activated signaling activities and characteristics of internalization. TRH-R1 and TRH-R2 are distributed differently in the brain and peripheral tIssues, but exhibit indistinguishable binding affinities for TRH and TRH analogs. Although they both can be stimulated by TRH to similar maximal signaling levels, TRH-R2 exhibits higher basal signaling activity and is more rapidly internalized than TRH-R1. These differences in signaling and internalization properties are probably important in the distinct parts that TRH-R1 and TRH-R2 may play in mammalian physiology.
- Published
- 2003
- Full Text
- View/download PDF
30. Impact of azaproline on amide cis-trans isomerism: conformational analyses and NMR studies of model peptides including TRH analogues.
- Author
-
Zhang WJ, Berglund A, Kao JL, Couty JP, Gershengorn MC, and Marshall GR
- Subjects
- Isomerism, Monte Carlo Method, Nuclear Magnetic Resonance, Biomolecular methods, Oligopeptides chemical synthesis, Oligopeptides chemistry, Proline chemistry, Protein Conformation, Receptors, Thyrotropin-Releasing Hormone chemistry, Receptors, Thyrotropin-Releasing Hormone metabolism, Thyrotropin-Releasing Hormone chemistry, Thyrotropin-Releasing Hormone metabolism, Aza Compounds chemistry, Proline analogs & derivatives, Thyrotropin-Releasing Hormone analogs & derivatives
- Abstract
The beta-turn is a well-studied motif in both proteins and peptides. Four residues, making almost a complete 180 degree-turn in the direction of the peptide chain, define the beta-turn. Several types of the beta-turn are defined according to Phi and Psi torsional angles of the backbone for residues i + 1 and i + 2. One special type of beta-turn, the type VI-turn, usually contains a proline with a cis-amide bond at residue i + 2. In an aza-amino acid, the alpha-carbon of the amino acid is changed to nitrogen. Peptides containing azaproline (azPro) have been shown to prefer the type VI beta-turn both in crystals and in organic solvents by NMR studies. MC/MD simulations using the GB/SA solvation model for water explored the conformational preferences of azPro-containing peptides in aqueous systems. An increase in the conformational preference for the cis-amide conformer of azPro was clearly seen, but the increased stability was relatively minor with respect to the trans-conformer as compared to previous suggestions. To test the validity of the calculations in view of the experimental data from crystal structures and NMR in organic solvents, [azPro(3)]-TRH and [Phe(2), azPro(3)]-TRH were synthesized, and their conformational preferences were determined by NMR in polar solvents as well as the impact of the azPro substitution on their biological activities.
- Published
- 2003
- Full Text
- View/download PDF
31. Homo- and hetero-oligomerization of thyrotropin-releasing hormone (TRH) receptor subtypes. Differential regulation of beta-arrestins 1 and 2.
- Author
-
Hanyaloglu AC, Seeber RM, Kohout TA, Lefkowitz RJ, and Eidne KA
- Subjects
- Animals, Arrestins chemistry, COS Cells, Cell Line, Chlorocebus aethiops, Cloning, Molecular, Fibroblasts metabolism, Genetic Vectors, Green Fluorescent Proteins, Kidney, Kinetics, Luminescent Proteins metabolism, Macromolecular Substances, Mice, Mice, Knockout, Polymerase Chain Reaction, Protein Isoforms chemistry, Rats, Receptors, Thyrotropin-Releasing Hormone metabolism, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins metabolism, Transfection, beta-Arrestin 1, beta-Arrestin 2, beta-Arrestins, Arrestins metabolism, Protein Isoforms metabolism, Receptors, Thyrotropin-Releasing Hormone chemistry
- Abstract
G-protein-coupled receptors (GPCRs) are regulated by a complex network of mechanisms such as oligomerization and internalization. Using the GPCR subtypes for thyrotropin-releasing hormone (TRHR1 and TRHR2), the aim of this study was to determine if subtype-specific differences exist in the trafficking process. If so, we wished to determine the impact of homo- and hetero-oligomerization on TRHR subtype trafficking as a potential mechanism for the differential cellular responses induced by TRH. Expression of either beta-arrestin 1 or 2 promoted TRHR1 internalization. In contrast, only beta-arrestin 2 could enhance TRHR2 internalization. The preference for beta-arrestin 2 by TRHR2 was supported by the impairment of TRHR2 trafficking in mouse embryonic fibroblasts (MEFs) from either a beta-arrestin 2 knockout or a beta-arrestin 1/2 knockout, while TRHR1 trafficking was only abolished in MEFs lacking both beta-arrestins. The differential beta-arrestin-dependence of TRHR2 was directly measured in live cells using bioluminescence resonance energy transfer (BRET). Both BRET and confocal microscopy were also used to demonstrate that TRHR subtypes form hetero-oligomers. In addition, these hetero-oligomers have altered internalization kinetics compared with the homo-oligomer. The formation of TRHR1/2 heteromeric complexes increased the interaction between TRHR2 and beta-arrestin 1. This may be due to conformational differences between TRHR1/2 hetero-oligomers versus TRHR2 homo-oligomers as a mutant TRHR1 (TRHR1 C335Stop) that does not interact with beta-arrestins, could also enhance TRHR2/beta-arrestin 1 interaction. This study demonstrates that TRHR subtypes are differentially regulated by the beta-arrestins and also provides the first evidence that the interactions of TRHRs with beta-arrestin may be altered by hetero-oligomer formation.
- Published
- 2002
- Full Text
- View/download PDF
32. Correlation between basal signaling and internalization of thyrotropin-releasing hormone receptors: evidence for involvement of similar receptor conformations.
- Author
-
Sun Y and Gershengorn MC
- Subjects
- Animals, Cells, Cultured, GTP-Binding Proteins physiology, Mice, Protein Conformation, Receptors, Cell Surface physiology, Receptors, Thyrotropin-Releasing Hormone chemistry, Receptors, Thyrotropin-Releasing Hormone metabolism
- Abstract
Previous studies have shown that rat thyrotropin-releasing hormone (TRH) receptor type 2 exhibits higher basal signaling activity and internalizes more rapidly upon agonist binding than rat TRH receptor type 1. The mouse TRH receptor type 2 (mR2) was recently cloned and, similar to its rat homolog, shows a higher basal signaling activity than mR1. Taking advantage of the high degree of sequence homology between mR1 and mR2, we used chimeras/mutants of these receptors to gain insight into the properties of the receptors that influence internalization and basal signaling. Chimeric receptors that have the mR1 extracellular and transmembrane domains with the carboxyl terminus and intracellular loops of mR2 (R1/R2-tail; R1/R2-I3,tail; R1/R2-I2,3,tail; R1/R2-I1,2,3,tail) exhibited internalization rates and basal activities that were similar to that of mR1. In contrast, a chimeric receptor with the extracellular and transmembrane domains of mR2 and the carboxyl terminus of mR1 exhibited the more rapid internalization rate and higher basal signaling activity characteristic of mR2. We showed previously that mutation of a highly conserved tryptophan to alanine caused mR1 to exhibit a high basal signaling activity and rapid internalization rate. In contrast, mutation of this tryptophan to alanine in mR2 decreased the rate of internalization and inhibited basal signaling activity. The rates of receptor internalization did not correlate with the binding affinities, coupling efficiencies, or potencies of the receptors. Thus, we observed that receptors with more rapid internalization rates showed relatively higher basal signaling activities, whereas receptors with lower basal signaling activities showed slower internalization rates. These data suggest that similar receptor conformations are required for productive coupling to signaling G proteins and to proteins involved in internalization.
- Published
- 2002
- Full Text
- View/download PDF
33. Constitutive and agonist-dependent homo-oligomerization of the thyrotropin-releasing hormone receptor. Detection in living cells using bioluminescence resonance energy transfer.
- Author
-
Kroeger KM, Hanyaloglu AC, Seeber RM, Miles LE, and Eidne KA
- Subjects
- Amino Acid Substitution, Animals, Arrestins metabolism, Bacterial Proteins analysis, Bacterial Proteins genetics, COS Cells, Cell Line, Cell Membrane physiology, Chlorocebus aethiops, Coated Pits, Cell-Membrane physiology, Energy Transfer, Humans, Iodine Radioisotopes, Luciferases analysis, Luciferases genetics, Luminescent Measurements, Luminescent Proteins analysis, Luminescent Proteins genetics, Macromolecular Substances, Mutagenesis, Site-Directed, Rats, Receptors, LHRH agonists, Receptors, LHRH chemistry, Receptors, LHRH physiology, Receptors, Thyrotropin-Releasing Hormone agonists, Recombinant Fusion Proteins agonists, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins metabolism, Transfection, Triptorelin Pamoate pharmacokinetics, beta-Arrestins, Receptors, Thyrotropin-Releasing Hormone chemistry, Receptors, Thyrotropin-Releasing Hormone physiology, Triptorelin Pamoate analogs & derivatives
- Abstract
The ability of G-protein-coupled receptors (GPCRs) to interact to form new functional structures, either forming oligomers with themselves or forming associations with other intracellular proteins, has important implications for the regulation of cellular events; however, little is known about how this occurs. Here, we have employed a newly emerging technology, bioluminescence resonance energy transfer (BRET), used to study protein-protein interactions in living cells, to demonstrate that the thyrotropin-releasing hormone receptor (TRHR) forms constitutive homo-oligomers. This formation of TRHR homo-oligomers in the absence of ligand was shown by demonstration of an energy transfer between TRHR molecules fused to either donor, Renilla luciferase (Rluc) or acceptor, enhanced yellow fluorescent protein (EYFP) molecules. This interaction was shown to be specific, since energy transfer was not detected between co-expressed tagged TRHRs and either complementary tagged gonadotropin-releasing hormone (GnRH) or beta(2)-adrenergic receptors. Furthermore, generation of a BRET signal between the TRHRs could only be inhibited by co-expression of the wild-type TRHR and not by other GPCRs. Agonist stimulation led to a time- and dose-dependent increase in the amount of energy transfer. Inhibition of receptor internalization by co-expression of dynamin mutant K44A did not affect the interaction between TRHRs, suggesting that clustering of receptors within clathrin-coated pits is not sufficient for energy transfer to occur. BRET also provided evidence for the agonist-induced oligomerization of another GPCR, the GnRH receptor (GnRHR), and the presence of an agonist-induced interaction of the adaptor protein, beta-arrestin, with TRHR and the absence of an interaction of beta-arrestin with GnRHR. This study supports the usefulness of BRET as a powerful tool for studying GPCR aggregations and receptor/protein interactions in general and presents evidence that the functioning unit of TRHRs exists as homomeric complexes.
- Published
- 2001
- Full Text
- View/download PDF
34. Analysis of the C-terminal tail of the rat thyrotropin-releasing hormone receptor-1 in interactions and cointernalization with beta-arrestin 1-green fluorescent protein.
- Author
-
Groarke DA, Drmota T, Bahia DS, Evans NA, Wilson S, and Milligan G
- Subjects
- Amino Acid Motifs, Amino Acid Sequence, Animals, Arrestins chemistry, Calcium metabolism, Cells, Cultured, Fluorescent Antibody Technique, Green Fluorescent Proteins, Humans, Indicators and Reagents chemistry, Indicators and Reagents metabolism, Luminescent Proteins chemistry, Luminescent Proteins metabolism, Molecular Sequence Data, Rats, Receptors, Thyrotropin-Releasing Hormone chemistry, Sequence Homology, Amino Acid, Transfection, beta-Arrestin 1, beta-Arrestins, Arrestins metabolism, Receptors, Thyrotropin-Releasing Hormone metabolism
- Abstract
Coexpression of the rat thyrotropin releasing hormone receptor-1 with beta-arrestin 1-green fluorescent protein (GFP) in human embryonic kidney 293 cells results in agonist-dependent translocation of the arrestin to the plasma membrane followed by its cointernalization with the receptor. Truncations of the receptor C-terminal tail from 93 to 50 amino acids did not alter this. Truncations to fewer than 47 amino acids prevented such interactions and inhibited but did not fully eliminate agonist-induced internalization of the receptor. Deletion and site-directed mutants of the C-terminal tail indicated that separate elimination of a potential casein kinase II phosphorylation site or clathrin/clathrin adapter motifs was insufficient to prevent either internalization of the receptor or its cointernalization with beta-arrestin 1-GFP. Alteration of sites of acylation reduced internalization and prevented interactions with beta-arrestin 1-GFP. Combinations of these mutants resulted in lack of interaction with beta-arrestin 1-GFP and a 10-fold reduction in internalization of the receptor. Despite this, the receptor construct that lacked the three protein sequence motifs was fully functional. These studies map sites that contribute the interactions of the thyrotropin releasing hormone receptor-1 C-terminal tail required for effective contacts with beta-arrestin 1-GFP and indicate key roles for these interactions in agonist-induced internalization of the receptor.
- Published
- 2001
- Full Text
- View/download PDF
35. Minireview: Insights into G protein-coupled receptor function using molecular models.
- Author
-
Gershengorn MC and Osman R
- Subjects
- Amino Acid Sequence, Animals, Binding Sites, Conserved Sequence, Humans, Ligands, Models, Molecular, Molecular Sequence Data, Protein Structure, Secondary, Receptors, Thyrotropin-Releasing Hormone chemistry, Receptors, Thyrotropin-Releasing Hormone physiology, Signal Transduction, Thyrotropin-Releasing Hormone chemistry, Thyrotropin-Releasing Hormone physiology, GTP-Binding Proteins physiology, Receptors, Cell Surface chemistry, Receptors, Cell Surface physiology
- Abstract
G protein-coupled receptors (GPCRs) represent the largest family of signal-transducing molecules known. They convey signals for light and many extracellular regulatory molecules. GPCRs have been found to be dysfunctional/dysregulated in a growing number of human diseases and have been estimated to be the targets of more than 30% of the drugs used in clinical medicine today. Thus, understanding how GPCRs function at the molecular level is an important goal of biological research. In order to understand function at this level, it is necessary to delineate the 3D structure of these receptors. Recently, the 3D structure of rhodopsin has been resolved, but in the absence of experimentally determined 3D structures of other GPCRs, a powerful approach is to construct a theoretical model for the receptor and refine it based on experimental results. Computer-generated models for many GPCRs have been constructed. In this article, we will review these studies. We will place the greatest emphasis on an iterative, bi-directional approach in which models are used to generate hypotheses that are tested by experimentation and the experimental findings are, in turn, used to refine the model. The success of this approach is due to the synergistic interaction between theory and experiment.
- Published
- 2001
- Full Text
- View/download PDF
36. Kinetic analysis of the internalization and recycling of [3H]TRH and C-terminal truncations of the long isoform of the rat thyrotropin-releasing hormone receptor-1.
- Author
-
Drmota T and Milligan G
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Cell Line, DNA Primers, Humans, Kinetics, Ligands, Molecular Sequence Data, Protein Isoforms chemistry, Rats, Receptors, Thyrotropin-Releasing Hormone chemistry, Tritium, Endocytosis, Protein Isoforms metabolism, Receptors, Thyrotropin-Releasing Hormone metabolism, Thyrotropin-Releasing Hormone metabolism
- Abstract
The C-terminal tail of the long splice variant of the rat thyrotropin-releasing hormone (TRH) receptor-1 (TRHR-1L) comprises around 93 amino acids. A series of C-terminal truncations was constructed and expressed transiently in HEK-293 cells. The extent of steady-state internalization of these in response to [(3)H]TRH was dependent upon the degree of truncation. Little effect was produced by deletion of the C-terminal to 50 amino acids, although there was a substantial decrease in the extent of internalization by deletion to 45-46 amino acids. The rate of internalization of TRHR-1L in response to ligand was substantially decreased by the acid-wash procedures often used in the analysis of cellular distribution of receptors with peptide ligands, and thus an alternative procedure using a Mes-containing buffer was employed in the present study. Apart from a truncation anticipated to eliminate post-translational acylation of the re-ceptor, which altered both the association and dissociation rates of [(3)H]TRH, the kinetics of ligand binding were unaffected by C-terminal truncation. Equally, the rate of recycling to the plasma membrane of internalized receptors was unaffected by C-terminal truncation. Although the extent of internalization of the full-length receptor was impaired by pre-exposure of cells to TRH, this was not true of C-terminal truncation mutants, which displayed limited steady-state internalization ratios. A mutant with a substantial C-terminal deletion also displayed decreased functional desensitization compared with the full-length receptor.
- Published
- 2000
37. Expression of thyrotropin-releasing hormone (TRH) receptor subtype 1 in mouse pancreatic islets and HIT-T15, an insulin-secreting clonal beta cell line.
- Author
-
Yamada M, Shibusawa N, Hashida T, Ozawa A, Monden T, Satoh T, and Mori M
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Blotting, Northern, Calcium metabolism, Cells, Cultured, Cricetinae, Gene Expression, Islets of Langerhans chemistry, Islets of Langerhans cytology, Islets of Langerhans drug effects, Mice, Mice, Inbred ICR, Molecular Sequence Data, Pituitary Gland chemistry, RNA isolation & purification, Receptors, Thyrotropin-Releasing Hormone biosynthesis, Receptors, Thyrotropin-Releasing Hormone chemistry, Reverse Transcriptase Polymerase Chain Reaction, Thyrotropin-Releasing Hormone pharmacology, Islets of Langerhans metabolism, RNA, Messenger biosynthesis, Receptors, Thyrotropin-Releasing Hormone genetics
- Abstract
Thyrotropin-releasing hormone (TRH), originally isolated as a hypothalamic hormone, has been reported to be present and released from the pancreatic beta cells, affecting pancreatic functions. However, it still remains unclear whether TRH receptor is expressed in the pancreas. In the present study, we characterized TRH receptors (TRHR) in mouse pancreatic islets and HIT-T15 cells, a hamster clonal beta cell line. RT-PCR study showed significant expression of TRHR subtype 1 (TRHR1) mRNA in both mouse pancreatic islets and HIT-T15 (HIT) cells. In contrast, there was no expression of TRHR2 mRNA, a novel subtype of TRHR which is expressed predominantly in the central nervous system. Sequencing analysis demonstrated that TRHR1 of the islets was identical to that in the pituitary, and cloned hamster TRHR1 shared 93.3 % homology with that of the mouse at the nucleic acid level. Northern blot analysis of TRHR 1 mRNA in HIT-T15 cells showed a single strong hybridization signal approximately 3.7 kb in length. Furthermore, Scatchard plot analysis in HIT-T15 cells revealed that the Kd value for MeTRH was 0.63 nM. Significant elevation of intracellular calcium concentration was observed in response to as little as 10 nM TRH , and this was not affected by removal of extracellular calcium. This is the first description indicating the presence of functional TRH receptor subtype 1 in the pancreatic beta cells, and our observations suggested the regulation of pancreatic function by TRH through autocrine or paracrine mechanisms.
- Published
- 2000
- Full Text
- View/download PDF
38. TRH-R2 exhibits similar binding and acute signaling but distinct regulation and anatomic distribution compared with TRH-R1.
- Author
-
O'Dowd BF, Lee DK, Huang W, Nguyen T, Cheng R, Liu Y, Wang B, Gershengorn MC, and George SR
- Subjects
- Amino Acid Sequence, Animals, COS Cells, Cloning, Molecular, Cyclic AMP Response Element-Binding Protein metabolism, DNA, Complementary metabolism, Dose-Response Relationship, Drug, Down-Regulation, In Situ Hybridization, Membrane Proteins chemistry, Molecular Sequence Data, Pituitary Gland metabolism, Protein Binding, Rats, Receptors, Thyrotropin-Releasing Hormone chemistry, Receptors, Thyrotropin-Releasing Hormone genetics, Sequence Homology, Amino Acid, Signal Transduction, Thyrotropin-Releasing Hormone metabolism, Thyrotropin-Releasing Hormone pharmacology, Time Factors, Transcription, Genetic, Brain metabolism, Membrane Proteins genetics, Membrane Proteins metabolism, Receptors, G-Protein-Coupled, Receptors, Thyrotropin-Releasing Hormone metabolism
- Abstract
TRH (thyroliberin) is a tripeptide (pGlu-His-ProNH2) that signals via G protein-coupled receptors. Until recently, only a single receptor for TRH was known (TRH-R1), but two groups identified a second receptor, TRH-R2. We independently discovered TRH-R2. Using an extensive set of TRH analogs, we found no differences in TRH-R1 and TRH-R2 binding or in acute stimulation of signaling. TRH-R2 was more rapidly internalized upon binding TRH and exhibited a greater level of TRH-induced down-regulation than TRH-R1. During prolonged exposure to TRH, cells expressing TRH-R2 exhibited a lower level of gene induction than cells expressing TRH-R1. TRH-R2 receptor mRNA was present in very discrete nuclei and regions of rat brain. A major mRNA transcript for TRH-R2 was seen in the cerebral cortex, pons, thalamus, hypothalamus, and midbrain with faint bands found in the striatum and pituitary. The extensive distribution of TRH-R2 in the brain suggests that it mediates many of the known functions of TRH that are not transduced by TRH-R1. The variations in agonist-induced internalization and down-regulation/desensitization, and anatomic distribution of TRH-R2 compared with TRH-R1, suggest important functional differences between the two receptors.
- Published
- 2000
- Full Text
- View/download PDF
39. A hydrophobic cluster between transmembrane helices 5 and 6 constrains the thyrotropin-releasing hormone receptor in an inactive conformation.
- Author
-
Colson AO, Perlman JH, Jinsi-Parimoo A, Nussenzveig DR, Osman R, and Gershengorn MC
- Subjects
- Animals, COS Cells, Cell Membrane chemistry, Computer Simulation, Luciferases metabolism, Midazolam pharmacology, Models, Molecular, Mutation, Phenylalanine chemistry, Plasmids, Protein Conformation, Receptors, Thyrotropin-Releasing Hormone agonists, Receptors, Thyrotropin-Releasing Hormone genetics, Transfection, Tyrosine chemistry, Cell Membrane metabolism, Receptors, Thyrotropin-Releasing Hormone chemistry, Tryptophan chemistry
- Abstract
We have studied the role of a highly conserved tryptophan and other aromatic residues of the thyrotropin-releasing hormone (TRH) receptor (TRH-R) that are predicted by computer modeling to form a hydrophobic cluster between transmembrane helix (TM)5 and TM6. The affinity of a mutant TRH-R, in which Trp279 was substituted by alanine (W279A TRH-R), for most tested agonists was higher than that of wild-type (WT) TRH-R, whereas its affinity for inverse agonists was diminished, suggesting that W279A TRH-R is constitutively active. We found that W279A TRH-R exhibited 3.9-fold more signaling activity than WT TRH-R in the absence of agonist. This increased basal activity was inhibited by the inverse agonist midazolam, confirming that the mutant receptor is constitutively active. Computer-simulated models of the unoccupied WT TRH-R, the TRH-occupied WT TRH-R, and various TRH-R mutants predict that a hydrophobic cluster of residues, including Trp279 (TM6), Tyr282, and Phe199 (TM5), constrains the receptor in an inactive conformation. In support of this model, we found that substitution of Phe199 by alanine or of Tyr282 by alanine or phenylalanine, but not of Tyr200 (by alanine or phenylalanine), resulted in a constitutively active receptor. We propose that a hydrophobic cluster including residues in TM5 and TM6 constrains the TRH-R in an inactive conformation via interhelical interactions. Disruption of these constraints by TRH binding or by mutation leads to changes in the relative positions of TM5 and TM6 and to the formation of an active form of TRH-R.
- Published
- 1998
- Full Text
- View/download PDF
40. Cloning and characterization of a cDNA encoding a novel subtype of rat thyrotropin-releasing hormone receptor.
- Author
-
Cao J, O'Donnell D, Vu H, Payza K, Pou C, Godbout C, Jakob A, Pelletier M, Lembo P, Ahmad S, and Walker P
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Calcium metabolism, Cloning, Molecular, DNA, Complementary metabolism, Humans, In Situ Hybridization, Mice, Molecular Sequence Data, Polymerase Chain Reaction, Radioligand Assay, Rats, Receptors, Thyrotropin-Releasing Hormone chemistry, Receptors, Thyrotropin-Releasing Hormone physiology, Recombinant Proteins biosynthesis, Recombinant Proteins chemistry, Sequence Alignment, Sequence Homology, Amino Acid, Thyrotropin-Releasing Hormone pharmacology, Thyrotropin-Releasing Hormone physiology, Transfection, Brain metabolism, Pituitary Gland metabolism, Receptors, Thyrotropin-Releasing Hormone genetics
- Abstract
A cDNA encoding a thyrotropin-releasing hormone (TRH) receptor expressed in the pituitary was previously cloned (De La Pena, P., Delgado, L. M., Del Camino, D., and Barros, F. (1992) Biochem. J. 284, 891-899; De La Pena, P., Delgado, L. M., Del Camino, D., and Barros, F. (1992) J. Biol. Chem. 267, 25703-25708; Duthie, S. M., Taylor, P. L., Anderson, J., Cook, J., and Eidne, K. A. (1993) Mol. Cell Endocrinol. 95, R11-R15). We now describe the isolation of a rat cDNA encoding a novel subtype of TRH receptor (termed TRHR2) displaying an overall homology of 50% to the pituitary TRH receptor. Introduction of TRHR2 cDNA in HEK-293 cells resulted in expression of high affinity TRH binding with a different pharmacological profile than the pituitary TRH receptor. De novo expressed receptors were functional and resulted in stimulation of calcium transient as assessed by fluorometric imaging plate reader analysis. The message for TRHR2 was exclusive to central nervous system tissues as judged by Northern blot analysis. Studies of the expression of TRHR-2 message by in situ hybridization revealed a pattern of expression remarkably distinct (present in spinothalamic tract, spinal cord dorsal horn) from that of the pituitary TRH receptor (present in hypothalamus, and ventral horn of the spinal cord, anterior pituitary). Therefore, we have identified a novel, pharmacologically distinct receptor for thyrotropin-releasing hormone that appears to be more restricted to the central nervous system particularly to the sensory neurons of spinothalamic tract and spinal cord dorsal horn, which may account for the sensory antinociceptive actions of TRH.
- Published
- 1998
- Full Text
- View/download PDF
41. User-friendly and versatile software for analysis of protein hydrophobicity.
- Author
-
Han B and Tashjian AH Jr
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Mathematical Computing, Molecular Sequence Data, Rats, Receptors, Thyrotropin-Releasing Hormone chemistry, Proteins chemistry, Software
- Abstract
We have developed a simple and flexible program to analyze regional hydrophobicity of a protein from its amino acid (aa) sequence. This program runs as a Microsoft Excel document into which aa sequence can be copied from any Windows-compatible or Macintosh word processor. The program returns the hydrophobicity index of each aa residue and other analyses that can be used to predict transmembrane domains, amphiphilic alpha helices and putative antigenic epitopes in a protein using established algorithms. The program can also be easily modified to test user-defined algorithms and to accommodate non-conventional aa residues or ambiguities in the aa sequence. Simple modification of the program allows direct use of nucleic acid sequence information for various analyses. Graphic visualization of the results is readily achieved using the graphics function of Microsoft Excel. Alternatively, the data can be imported into other graphics software for preparation of publication-quality figures. By running as a document in Microsoft Excel, which can be found in virtually all personal computers, this program provides easy access even to the computer novice.
- Published
- 1998
- Full Text
- View/download PDF
42. Cloning and characterization of the chicken thyrotropin-releasing hormone receptor.
- Author
-
Sun YM, Millar RP, Ho H, Gershengorn MC, and Illing N
- Subjects
- Amino Acid Sequence, Animals, Binding Sites, COS Cells, DNA, Complementary chemistry, DNA, Complementary genetics, Mice, Molecular Sequence Data, Receptors, Thyrotropin-Releasing Hormone chemistry, Sequence Analysis, DNA, Transfection, Chickens genetics, Cloning, Molecular, Receptors, Thyrotropin-Releasing Hormone genetics
- Abstract
We report on the cloning of the full-length complementary DNA for the chicken TRH receptor. Although the TRH receptor has been cloned from several mammalian species, this is the first report from another vertebrate class. The ligand binding pocket, which is situated in the transmembrane helices of the mouse and rat TRH receptors, is completely conserved in the chicken receptor. Pharmacological studies (receptor binding and signaling) employing several TRH analogs revealed that there are no significant differences between the chicken and mouse receptors. These findings show that there have been considerable evolutionary constraints on TRH receptor structure and function. Several truncated forms of the chicken TRH receptor that appear to retain a part of an intron and are truncated in the putative third intracellular loop were also cloned, but were nonfunctional. This study provides a useful tool for further studies on the roles of TRH in avian growth and TSH regulation.
- Published
- 1998
- Full Text
- View/download PDF
43. Static and dynamic roles of extracellular loops in G-protein-coupled receptors: a mechanism for sequential binding of thyrotropin-releasing hormone to its receptor.
- Author
-
Colson AO, Perlman JH, Smolyar A, Gershengorn MC, and Osman R
- Subjects
- Amino Acid Sequence, Animals, Binding Sites, COS Cells, Cloning, Molecular, Computer Simulation, Conserved Sequence, GTP-Binding Proteins metabolism, Mice, Models, Molecular, Molecular Sequence Data, Mutagenesis, Site-Directed, Recombinant Proteins chemistry, Recombinant Proteins metabolism, Software, Thyrotropin-Releasing Hormone chemistry, Transfection, Protein Structure, Secondary, Receptors, Thyrotropin-Releasing Hormone chemistry, Receptors, Thyrotropin-Releasing Hormone metabolism, Thyrotropin-Releasing Hormone metabolism
- Abstract
Small ligands generally bind within the seven transmembrane-spanning helices of G-protein-coupled receptors, but their access to the binding pocket through the closely packed loops has not been elucidated. In this work, a model of the extracellular loops of the thyrotropin-releasing hormone (TRH) receptor (TRHR) was constructed, and molecular dynamics simulations and quasi-harmonic analysis have been performed to study the static and dynamic roles of the extracellular domain. The static analysis based on curvature and electrostatic potential on the surface of TRHR suggests the formation of an initial recognition site between TRH and the surface of its receptor. These results are supported by experimental evidence. A quasi-harmonic analysis of the vibrations of the extracellular loops suggest that the low-frequency motions of the loops will aid the ligand to access its transmembrane binding pocket. We suggest that all small ligands may bind sequentially to the transmembrane pocket by first interacting with the surface binding site and then may be guided into the transmembrane binding pocket by fluctuations in the extracellular loops.
- Published
- 1998
- Full Text
- View/download PDF
44. Role of the extracellular loops of the thyrotropin-releasing hormone receptor: evidence for an initial interaction with thyrotropin-releasing hormone.
- Author
-
Perlman JH, Colson AO, Jain R, Czyzewski B, Cohen LA, Osman R, and Gershengorn MC
- Subjects
- Animals, Binding Sites, COS Cells, Computer Simulation, GTP-Binding Proteins metabolism, Hydrogen Bonding, Kinetics, Ligands, Mice, Models, Molecular, Mutagenesis, Protein Binding, Protein Conformation, Protein Structure, Secondary, Receptors, Thyrotropin-Releasing Hormone genetics, Transfection genetics, Tyrosine chemistry, Tyrosine metabolism, Receptors, Thyrotropin-Releasing Hormone chemistry, Receptors, Thyrotropin-Releasing Hormone metabolism, Thyrotropin-Releasing Hormone metabolism
- Abstract
Thyrotropin-releasing hormone (TRH), like most small ligands, appears to bind within the seven transmembrane-spanning helices (TMs) of its G protein-coupled receptor (TRH-R). A role for the extracellular loops (ECLs) of TRH-R has not been established. We substituted residues in the ECLs of TRH-R and show that Tyr-181 is important for high-affinity binding because its substitution leads to a 3700-fold lowering of the estimated affinity compared to wild-type TRH-R. Using TRH analogues, we provide evidence that there is a specific interaction between Tyr-181 in ECL-2 and the pyroGlu moiety of TRH. It was previously suggested that the pyroGlu of TRH may interact with Asn-110 in TM-3 and with Asn-289 in ECL-3; N110A and N289A TRH-Rs exhibit similar apparent affinities that are only 20-30-fold lower than wild-type TRH-R. To better understand these findings, we analyzed a computer-generated model which predicts that the ECLs form an entry channel into the TRH-R TM bundle, that Tyr-181 projects into this channel and that the pyroGlu of TRH cannot simultaneously interact with residues in the TMs and ECLs. Kinetic analysis showed that the association rate of [Ntau-methyl-His]TRH with N289A TRH-R is slower than with wild-type TRH-R and largely accounts for the lower apparent affinity; the association rate with N110A TRH-R is similar to that of wild-type TRH-R. These data are consistent with the idea that there are initial interactions between TRH and the residues of a putative entry channel of TRH-R. We suggest that a role of the ECLs in all G protein-coupled receptors for small ligands may be to initially contact the ligand and allow entry into a TM binding pocket.
- Published
- 1997
- Full Text
- View/download PDF
45. Interactions between conserved residues in transmembrane helices 1, 2, and 7 of the thyrotropin-releasing hormone receptor.
- Author
-
Perlman JH, Colson AO, Wang W, Bence K, Osman R, and Gershengorn MC
- Subjects
- Amino Acid Sequence, Animals, Asparagine, Aspartic Acid, Binding Sites, COS Cells, Computer Simulation, Conserved Sequence, Inositol Phosphates metabolism, Models, Structural, Mutagenesis, Site-Directed, Point Mutation, Recombinant Proteins chemistry, Recombinant Proteins metabolism, Thermodynamics, Thyrotropin-Releasing Hormone pharmacology, Transfection, Protein Structure, Secondary, Receptors, Thyrotropin-Releasing Hormone chemistry, Receptors, Thyrotropin-Releasing Hormone metabolism, Thyrotropin-Releasing Hormone chemistry, Thyrotropin-Releasing Hormone metabolism
- Abstract
The roles of conserved residues in transmembrane helices (TMs) of G protein-coupled receptors have not been well established. A computer-generated model of the thyrotropin-releasing hormone receptor (TRH-R) indicated that conserved Asp-71 (TM-2) could interact with conserved asparagines 316 (TM-7) and 43 (TM-1). To test this model, we constructed mutant TRH-Rs containing polar or alanine substitutions of these residues. The maximal activities of N43A and N316A TRH-Rs were diminished, whereas D71A (Perlman, J. H., Nussenzveig, D. R., Osman, R., and Gershengorn, M. C. (1992) J. Biol. Chem. 267, 24413-24417) and N43A/N316A TRH-Rs were inactive. Computer models of D71A and N43A/N316A TRH-Rs show similar changes from native TRH-R in their TM bundle conformations. The inactivity and the similarity of the computer models of D71A and N43A/N316A TRH-Rs are consistent with the idea that Asp-71 bridges Asn-43 and Asn-316 and suggest that activity is critically dependent on these interactions. The conservation of these residues suggests these specific interactions involving TMs 1, 2, and 7 may be structurally important for all members of the rhodopsin/beta-adrenergic receptor subfamily of G protein-coupled receptors.
- Published
- 1997
- Full Text
- View/download PDF
46. Altered ligand dissociation rates in thyrotropin-releasing hormone receptors mutated in glutamine 105 of transmembrane helix III.
- Author
-
del Camino D, Barros F, Pardo LA, and de la Peña P
- Subjects
- Amino Acid Sequence, Animals, Aspartic Acid, COS Cells, Conserved Sequence, Female, Hormones chemistry, Kinetics, Ligands, Molecular Sequence Data, Mutagenesis, Site-Directed, Oocytes physiology, Receptors, Thyrotropin-Releasing Hormone physiology, Recombinant Proteins chemistry, Recombinant Proteins metabolism, Sequence Homology, Amino Acid, Thyrotropin-Releasing Hormone metabolism, Thyrotropin-Releasing Hormone pharmacology, Transfection, Glutamine, Point Mutation, Protein Structure, Secondary, Receptors, Cell Surface chemistry, Receptors, Thyrotropin-Releasing Hormone chemistry, Receptors, Thyrotropin-Releasing Hormone metabolism, Thyrotropin-Releasing Hormone chemistry
- Abstract
Glutamine 105 in the third transmembrane helix of the thyrotropin-releasing hormone receptor (TRH-R) occupies a position equivalent to a conserved negatively charged residue in receptors for biogenic amines where it acts as counterion interacting with the cationic amine moiety of the ligand. Maximum levels of response to TRH in oocytes expressing wild-type TRH-Rs were indistinguishable from those of oocytes expressing receptors mutated to Glu, Asn, or Asp in position 105. However, the EC50 values for activation of oocyte responses increased more than 500 times in oocytes expressing mutant Glu105 receptors, in which the amido group of Gln105 has been removed by site-directed mutagenesis. Charge effects do not seem to be involved in the huge effect of mutating Gln105 to Glu, since mutation of Gln105 to Asp induces only a 15-fold increase in EC50. Furthermore, no change in EC50 is observed after mutation of Asn110 to Asp. The affinity shift (identified by changes in EC50 values for systems of comparable efficacy) in Glu105 mutant receptors was partially recovered in oocytes expressing Asn105 mutant receptors. These results and those obtained after substitution of Lys, Leu, Tyr, and Ser for Gln105 suggest that the presence and the correct position of the Gln hydrogen bond-donor amido group are important for normal functionality of the receptor. In wild type or Asp105 mutant receptors showing the same maximal responses, decreases in affinity with TRH and methyl-histidyl-TRH correlated with increased dissociation rates of hormone from the receptor. Rapid dilution experiments following subsecond stimulation indicate that the TRH-R is converted rapidly from a form showing fast dissociation kinetics to a form from which the hormone dissociates slowly. Mutation of residue 105 impairs the receptor shift between these two forms. This effect was demonstrated in a direct way by comparing [3H]methyl-histidyl-TRH dissociation rates in COS-7 cells transfected with either wild type or Asp105 mutant TRH-Rs. Thus, residues located in transmembrane helix III positions equivalent to those of the counterions for biogenic amines, regulate hormone-receptor interactions in the TRH receptor (and perhaps other receptors). Furthermore, the nature of the amino acid in these positions may also play a role, directly or indirectly, in conformational changes leading to receptor activation, and hence to signal transduction.
- Published
- 1997
- Full Text
- View/download PDF
47. A disulfide bonding interaction role for cysteines in the extracellular domain of the thyrotropin-releasing hormone receptor.
- Author
-
Cook JV, McGregor A, Lee T, Milligan G, and Eidne KA
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Binding Sites, Binding, Competitive, Cell Line, Cell Membrane metabolism, Chlorocebus aethiops, DNA Primers, Disulfides, GTP-Binding Proteins metabolism, Humans, Kinetics, Molecular Sequence Data, Mutagenesis, Insertional, Mutagenesis, Site-Directed, Pituitary Gland, Anterior metabolism, Point Mutation, Polymerase Chain Reaction, Protein Structure, Secondary, Rats, Recombinant Proteins chemistry, Recombinant Proteins metabolism, Thyrotropin-Releasing Hormone metabolism, Thyrotropin-Releasing Hormone pharmacology, Transfection, Cysteine, Dithiothreitol pharmacology, Receptors, Thyrotropin-Releasing Hormone chemistry, Receptors, Thyrotropin-Releasing Hormone metabolism
- Abstract
The roles of disulfide and sulfhydryl groups in the specific binding of TRH to its receptor have been examined. In all TRH receptors (TRH-Rs) isolated from different species so far, there are only two extracellular cysteine residues (Cys98 in the extracellular loop between transmembrane helices 2 and 3 and Cys179 in the extracellular loop between transmembrane helices 4 and 5) that are in positions homologous to cysteine residues in other G protein-coupled receptors. Another Cys (Cys100) is located in close proximity to Cys98 at the interface between the first extracellular loop and third transmembrane domain. To assess the role of these TRH-R Cys residues in disulfide bonding interactions, they were mutated to either Ser or Ala. Six mutant receptors (Cys98Ser, Cys98Ala, Cys179Ser, Cys179Ala, Cys100Ser, and Cys100Ala) were expressed in COS-1 cells and tested for their ability to bind TRH and to activate total inositol phosphate (IP) formation. TRH-R mutants Cys100Ser and Cys100Ala showed TRH binding affinities and IP activation similar to the wild-type (WT). In contrast, mutants Cys98Ser, Cys98Ala, Cys179Ser, and Cys179Ala showed no high affinity TRH binding. The potencies of Cys98Ala and Cysl79Ala as measured by IP stimulation were decreased by four orders of magnitude when compared with WT. Cys98Ser potency decreased by five orders of magnitude, whereas Cys179Ser showed no IP production. Northern blotting confirmed expression of all the mutant TRH-Rs at the messenger RNA (mRNA) level. An epitope tag derived from the Haemophilus influenza hemagglutinin protein was incorporated at the NH2 termini of the TRH-R WT and TRH-R Cys mutants to allow the independent assessment of cell surface expression of receptor protein. TRH-R mutants that failed to show receptor binding (Cys98Ser, Cys98Ala, Cys179Ala) showed WT levels of cell surface receptor expression, indicating that loss of receptor binding in these mutants is not attributable to loss of receptor expression. In contrast, cell surface expression of Cysl79Ser, which showed no ligand induced IP stimulation, could not be detected. Dithiothreitol, a disulfide bond reducing agent, and p-chloromercuribenzoic acid (p-CMB), a sulfhydryl blocking compound, reduced specific TRH binding in a dose-dependent manner. The inhibition of binding by dithiothreitol implies that the integrity of a disulfide bond is important for TRH binding to its receptor. The dramatic inhibition of TRH binding by p-CMB indicates that free sulfhydryl groups are also associated with the binding of the ligand to its receptor. This study presents evidence that a disulfide bond exists between Cys98 and Cys179 which is essential for maintaining the receptor in the correct conformation for ligand binding. Cys100 is not thought to have a disulfide bonding interaction role. Results obtained after chemical modification have shown that free sulfhydryl groups within the TRH-R may also have a role in ligand interactions.
- Published
- 1996
- Full Text
- View/download PDF
48. A refined model of the thyrotropin-releasing hormone (TRH) receptor binding pocket. Novel mixed mode Monte Carlo/stochastic dynamics simulations of the complex between TRH and TRH receptor.
- Author
-
Laakkonen LJ, Guarnieri F, Perlman JH, Gershengorn MC, and Osman R
- Subjects
- Amino Acid Sequence, Animals, Calorimetry, Computer Simulation, Mice, Models, Molecular, Models, Structural, Molecular Sequence Data, Monte Carlo Method, Software, Stochastic Processes, Protein Conformation, Protein Structure, Secondary, Receptors, Thyrotropin-Releasing Hormone chemistry, Receptors, Thyrotropin-Releasing Hormone metabolism, Thyrotropin-Releasing Hormone chemistry, Thyrotropin-Releasing Hormone metabolism
- Abstract
Previous mutational and computational studies of the thyrotropin-releasing hormone (TRH) receptor identified several residues in its binding pocket [see accompanying paper, Perlman et al. (1996) Biochemistry 35, 7643-7650]. On the basis of the initial model constructed with standard energy minimization techniques, we have conducted 15 mixed mode Monte Carlo/stochastic dynamics (MC-SD) simulations to allow for extended sampling of the conformational states of the ligand and the receptor in the complex. A simulated annealing protocol was adopted in which the complex was cooled from 600 to 310 K in segments of 30 ps of the MC-SD simulations for each change of 100 K. Analysis of the simulation results demonstrated that the mixed mode MC-SD protocol maintained the desired temperature in the constant temperature simulation segments. The elevated temperature and the repeating simulations allowed for adequate sampling of the torsional space of the complex with successful conservation of the general structure and good helicity of the receptor. For the analysis of the interaction between TRH and the binding pocket, TRH was divided into four groups consisting of pyroGlu, His, ProNH2, and the backbone. The pairwise interaction energies of the four separate portions of TRH with the corresponding residues in the receptor provide a physicochemical basis for the understanding of ligand-receptor complexes. The interaction of pyroGlu with Tyr106 shows a bimodal distribution that represents two populations: one with a H-bond and another without it. Asp195 was shown to compete with pyroGlu for the H-bond to Tyr106. Simulations in which Asp195 was interacting with Arg283, thus removing it from the vicinity of Tyr106, resulted in a stable H-bond to pyroGlu. In all simulations His showed a van der Waals attraction to Tyr282 and a weak electrostatic repulsion from Arg 306. The ProNH2 had a strong and frequent H-bonding interaction with Arg306. The backbone carbonyls show a frequent H-bonding interaction with the OH group of Tyr282 and strong, often multiple, interactions with Arg306. Three structures, which maintained these interactions simultaneously, were selected as candidates for ligand-receptor complexes. These show persistent interactions of TRH with Ile 109 and Ile 116 in HX 3 and with Tyr310 and Ser313 in HX 7, which will be tested to refine the structure of the ligand-receptor complex. The superposition of the three structures shows the extent of structural flexibility of the receptor and the ligand in the complex. The backbone of TRH inside the receptor is in an alpha-helical conformation, suggesting that the receptor, through its interaction with the ligand, provides the energy required for the conformational change in the ligand from an extended to the folded form.
- Published
- 1996
- Full Text
- View/download PDF
49. A refined model of the thyrotropin-releasing hormone (TRH) receptor binding pocket. Experimental analysis and energy minimization of the complex between TRH and TRH receptor.
- Author
-
Perlman JH, Laakkonen LJ, Guarnieri F, Osman R, and Gershengorn MC
- Subjects
- Amino Acid Sequence, Animals, Binding Sites, Chlorocebus aethiops, Kidney, Kinetics, Mathematics, Mice, Molecular Sequence Data, Monte Carlo Method, Mutagenesis, Site-Directed, Point Mutation, Polymerase Chain Reaction, Recombinant Proteins chemistry, Recombinant Proteins metabolism, Stochastic Processes, Thyrotropin-Releasing Hormone analogs & derivatives, Transfection, Models, Molecular, Protein Conformation, Protein Structure, Secondary, Receptors, Thyrotropin-Releasing Hormone chemistry, Receptors, Thyrotropin-Releasing Hormone metabolism, Thyrotropin-Releasing Hormone chemistry, Thyrotropin-Releasing Hormone metabolism
- Abstract
Seven transmembrane (TM) spanning, G protein-coupled receptors (GPCRs) appear to bind large glycoprotein hormones predominantly within their extracellular domains, small nonpeptidic ligands within the TM helical bundle, and peptide ligands within the extracellular domains and TM bundle. The tripeptide thyrotropin-releasing hormone (TRH, pyroGlu-His-ProNH2) may bind entirely within the TM bundle of the TRH receptor (TRH-R). We have previously demonstrated direct binding contacts between the pyroGlu of TRH and two residues in TM helix 3 (TM-3) of TRH-R and proposed a model of the binding pocket of TRH-R [Perlman, J. H., Laakkonen, L., Osman, R., & Gershengorn, M. C. (1994) J. Biol. Chem. 269, 23383-23386]. Here, we provide evidence for two additional direct interactions between TRH and TRH-R. One interaction is between the aromatic ring of Tyr 282 of TM-6 and His of TRH. This is based on a large increase in the half-maximally effective concentration (EC50) of TRH for stimulation of inositol phosphate formation by Y282A TRH-R and a loss of selectivity of this mutant receptor for TRH analogs substituted at His. We provide evidence for another interaction between Arg 306 of TM-7 and the terminal carboxamide of TRH. Using four direct interactions as anchors, a refined model of the TRH-R binding pocket was constructed using geometry optimization through energy minimization. A novel method for modeling GPCRs based on Monte Carlo and stochastic dynamics simulations is presented in the accompanying paper [Laakkonen, L. J., Guarnieri, F., Perlman, J. H., Gershengorn, M. C., & Osman, R. (1996) Biochemistry 35, 7651-7663].
- Published
- 1996
- Full Text
- View/download PDF
50. Restricted analogues provide evidence of a biologically active conformation of thyrotropin-releasing hormone.
- Author
-
Laakkonen L, Li W, Perlman JH, Guarnieri F, Osman R, Moeller KD, and Gershengorn MC
- Subjects
- Amino Acid Sequence, Animals, Mice, Molecular Sequence Data, Protein Conformation, Receptors, Thyrotropin-Releasing Hormone chemistry, Structure-Activity Relationship, Thyrotropin-Releasing Hormone pharmacology, Thyrotropin-Releasing Hormone chemistry
- Abstract
Thyrotropin-releasing hormone (TRH) is a tripeptide (< Glu-His-Pro-NH2) that signals through a G protein-coupled receptor. TRH is a highly flexible molecule that can assume many conformations in solution. To attempt to delineate the biologically active conformation of TRH, we synthesized a pair of conformationally restricted cyclohexyl/Ala2-TRH analogues. The diastereomeric analogues use a lactam ring to restrict two of the six free torsional angles of TRH and constrain the X-Pro-NH2 peptide bond to trans. Unrestricted cyclohexyl/Ala2-TRH exhibited a 650-fold lower affinity than TRH for TRH receptor and was 430-fold less potent than TRH in stimulating inositol phosphate second messenger formation. One diastereomer exhibited higher affinity and potency than the unrestricted analogue despite the presence of the methylene bridge and fused ring, whereas the other showed lower affinity and potency. Computer simulations predicted that the positions of the cyclohexyl/Ala2 and Pro-NH2 moieties relative to < glutamate were different in the two analogues and that the conformation of the higher affinity analogue is different from that of trans-TRH in solution but is superimposable on that of trans-TRH found in a model of the TRH/TRH receptor complex. These experimental findings identify a favored relative position of < glutamate and Pro-NH2 in the more active conformation of two diastereomeric analogues of TRH and provide independent support for the model of the TRH/TRH receptor complex.
- Published
- 1996
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.