Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 166
Filter
1.
Horm Metab Res ; 47(10): 789-96, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26197854

ABSTRACT

Although there are adequate therapies for Graves' hyperthyroidism, mild to moderate Graves' orbitopathy (GO) is usually treated symptomatically whereas definitive therapy is reserved for severe, vision-threatening GO. Importantly, none of the treatment regimens for Graves' disease used today are directed at the pathogenesis of the disease. Herein, we review some aspects of what is known about the pathogenesis of these 2 major components of Graves' disease, specifically the apparent important roles of the TSH and IGF-1 receptors, and thereafter describe future therapeutic approaches directed at these receptors. We propose that targeting these receptors will yield effective and better tolerated treatments for Graves' disease, especially for GO.


Subject(s)
Graves Ophthalmopathy/therapy , Autoantibodies/immunology , Humans , Molecular Targeted Therapy , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/metabolism , Receptors, Thyrotropin/antagonists & inhibitors , Receptors, Thyrotropin/immunology , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use
2.
Ann Endocrinol (Paris) ; 72(2): 74-6, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21511239

ABSTRACT

TSH activates the TSH receptor (TSHR) thereby stimulating the function of thyroid follicular cells (thyrocytes) leading to biosynthesis and secretion of thyroid hormones. Because TSHR is involved in several thyroid pathologies, there is a strong rationale for the design of small molecule "drug-like" ligands. Recombinant human TSH (rhTSH, Thyrogen(®)) has been used in the follow-up of patients with thyroid cancer to increase the sensitivity for detection of recurrence or metastasis. rhTSH is difficult to produce and must be administered by injection. A small molecule TSHR agonist could produce the same beneficial effects as rhTSH but with greater ease of oral administration. We developed a small molecule ligand that is a full agonist at TSHR. Importantly for its clinical potential, this agonist elevated serum thyroxine and stimulated thyroidal radioiodide uptake in mice after its absorption from the gastrointestinal tract following oral administration. Graves' disease (GD) is caused by persistent, unregulated stimulation of thyrocytes by thyroid-stimulating antibodies (TSAbs) that activate TSHR. We identified the first small molecule TSHR antagonists that inhibited TSH- and TSAb-stimulated signalling in primary cultures of human thyrocytes. Our results provide proof-of-principle for effectiveness of small molecule agonists and antagonists for TSHR. We suggest that these small molecule ligands are lead compounds for the development of higher potency ligands that can be used as probes of TSHR biology with therapeutic potential.


Subject(s)
Drug Discovery , Graves Disease/drug therapy , Receptors, Thyrotropin/agonists , Receptors, Thyrotropin/antagonists & inhibitors , Thyroid Neoplasms/drug therapy , Animals , Cells, Cultured , Drug Evaluation, Preclinical , Humans , Iodine Isotopes/metabolism , Ligands , Mice , Signal Transduction/drug effects , Small Molecule Libraries , Thyroid Function Tests , Thyroid Gland/drug effects , Thyroxine/blood
3.
Cell Prolif ; 41(3): 474-91, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18422699

ABSTRACT

OBJECTIVES: Previously, we characterized human islet-derived precursor cells (hIPCs) as mesenchymal stem cells that migrate out from islets in vitro and can differentiate into functional islet-like structures following proliferative expansion. Here, we investigate the role of beta-catenin signalling in derivation and proliferation of hIPCs. MATERIALS AND METHODS: Localization of beta-catenin was performed using confocal microscopy. Expression levels of beta-catenin target genes were measured by quantitative real-time polymerase chain reaction. Loss-of-function studies were performed using specific short interfering RNAs. RESULTS: Immunostaining of islet outgrowths revealed translocation of beta-catenin from plasma membranes in intact islets to the nucleus in cells migrating out. There were no nuclear beta-catenin-positive cells in intact islets whereas between 35% and 70% of cells in established hIPC cultures exhibited nuclear beta-catenin. Transcripts for beta-catenin target genes were increased in hIPCs compared to those in islets. Beta-catenin translocated to the cell membrane when hIPCs formed epithelial cell clusters. In proliferating hIPCs, there was a strong correlation between markers of proliferation and nuclear beta-catenin. Treatment of hIPCs with the glycogen synthase kinase-3beta inhibitor (2'Z,3'E)-6-Bromoindirubin-3'-oxime increased intracellular beta-catenin but reduced nuclear beta-catenin, and was associated with reduced cell proliferation. Finally, knockdown of beta-catenin decreased beta-catenin target gene expression and hIPC proliferation. CONCLUSIONS: These results support a functional role for beta-catenin during proliferation of hIPCs and suggest that activated beta-catenin signalling may also be important during hIPC derivation from islets.


Subject(s)
Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Mesoderm/cytology , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , beta Catenin/metabolism , Biomarkers/metabolism , Cell Nucleus/metabolism , Cell Proliferation , Epithelium/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Mesoderm/metabolism , Protein Transport , Transcription, Genetic , Wnt Proteins/genetics
4.
Mini Rev Med Chem ; 6(2): 221-6, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16472189

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.


Subject(s)
Thyrotropin-Releasing Hormone/analogs & derivatives , Animals , Central Nervous System/drug effects , Humans , Receptors, Thyrotropin-Releasing Hormone/chemistry , Thyrotropin-Releasing Hormone/pharmacology
5.
J Mol Endocrinol ; 30(2): 87-97, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12683933

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.


Subject(s)
Protein Isoforms/metabolism , Receptors, Thyrotropin-Releasing Hormone/metabolism , Signal Transduction/physiology , Thyrotropin-Releasing Hormone/metabolism , 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
6.
J Biol Chem ; 276(36): 33805-11, 2001 Sep 07.
Article in English | MEDLINE | ID: mdl-11448967

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV; human herpesvirus 8) encodes a chemokine-like G protein-coupled receptor (KSHV-GPCR) that is implicated in the pathogenesis of Kaposi's sarcoma (KS). Since endothelial cells appear to be targets for the virus, we developed an in vitro mouse lung endothelial cell model in which KSHV-GPCR is stably expressed and KSHV-GPCR signaling was studied. In mouse lung endothelial cells: 1) KSHV-GPCR does not exhibit basal signaling through the phosphoinositide-specific phospholipase C pathway but inositol phosphate production is stimulated by growth-related oncogene alpha (Gro-alpha) via a pertussis toxin (PTX)-insensitive pathway; 2) KSHV-GPCR signals basally through a PTX-sensitive pathway leading to a lowering of intracellular cAMP level that can be lowered further by Gro alpha and increased by interferon gamma-inducible protein 10; 3) KSHV-GPCR stimulates phosphatidylinositol 3-kinase via a PTX-insensitive mechanism; and 4) KSHV-GPCR activates nuclear factor-kappa B (NF-kappa B) by a PTX-sensitive G beta gamma subunit-mediated pathway. These data show that KSHV-GPCR couples to at least two G proteins and initiates signaling via at least three cascades in endothelial cells thereby increasing the complexity of regulation of endothelial cell function by KSHV-GPCR that may occur during viral infection.


Subject(s)
Endothelium/metabolism , Receptors, Chemokine/metabolism , Adenylyl Cyclases/metabolism , Animals , Animals, Newborn , Binding, Competitive , Cell Survival , Colforsin/metabolism , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Fibroblasts/metabolism , Luciferases/metabolism , Lung/metabolism , Mice , NF-kappa B/metabolism , Pertussis Toxin , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding , Signal Transduction , Time Factors , Transfection , Type C Phospholipases/metabolism , Virulence Factors, Bordetella/pharmacology
7.
Endocrinology ; 142(3): 1188-94, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11181534

ABSTRACT

We cloned the mouse TRH receptor type 2 (mTRH-R2) gene, which is 92% identical with rat TRH-R2 and 50% identical with mTRH-R1 at the amino acid level, and identified an intron within the coding sequence that is not present in the TRH-R1 gene structure. Similar to its rat homolog, mTRH-R2 binds TRH with an affinity indistinguishable from mTRH-R1, signals via the phosphoinositide pathway like mTRH-R1, but exhibits a higher basal signaling activity than mTRH-R1. We found that regulator of G protein signaling 4 (RGS4), which differentially inhibits signaling by other receptors that couple to Gq, inhibits TRH-stimulated signaling via mTRH-R1 and mTRH-R2 to similar extents. In contrast, other RGS proteins including RGS7, RGS9, and GAIP had no effect on signaling by mTRH-R1 or mTRH-R2 demonstrating the specificity of RGS4 action. Interestingly, RGS4 markedly inhibited basal signaling by mTRH-R2. Inhibition of basal signaling of mTRH-R2 by RGS4 suggests that modulation of agonist-independent signaling may be an important mechanism of regulation of G protein-coupled receptor activity under normal physiologic circumstances.


Subject(s)
RGS Proteins/pharmacology , Receptors, Thyrotropin-Releasing Hormone/physiology , Signal Transduction/drug effects , Thyrotropin-Releasing Hormone/pharmacology , Amino Acid Sequence/genetics , Animals , Blotting, Northern , Cell Line , Cloning, Molecular , Humans , Mice , Molecular Sequence Data , Protein Isoforms/genetics , Protein Isoforms/physiology , Receptors, Thyrotropin-Releasing Hormone/genetics
8.
Endocrinology ; 142(1): 2-10, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11145559

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.


Subject(s)
GTP-Binding Proteins/physiology , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/physiology , 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
9.
J Biol Chem ; 276(2): 1376-82, 2001 Jan 12.
Article in English | MEDLINE | ID: mdl-11031271

ABSTRACT

Because charged residues at the intracellular ends of transmembrane helix (TMH) 2 and TMH3 of G protein-coupled receptors (GPCRs) affect signaling, we performed mutational analysis of these residues in the constitutively signaling Kaposi's sarcoma-associated herpesvirus GPCR (KSHV-GPCR). KSHV-GPCR contains the amino acid sequence Val-Arg-Tyr rather than the Asp/Glu-Arg-Tyr ((D/E)RY) motif at the intracellular end of TMH3. Mutation of Arg-143 to Ala (R143A) or Gln (R143Q) abolished constitutive signaling whereas R143K exhibited 50% of the basal activity of KSHV-GPCR. R143A was not stimulated by agonist, whereas R143Q was stimulated by growth-related oncogene-alpha, and R143K, similar to KSHV-GPCR, was stimulated further. These findings show that Arg-143 is critical for signal generation in KSHV-GPCR. In other GPCRs, Arg in this position may act as a signaling switch by movement of its sidechain from a hydrophilic pocket in the TMH bundle to a position outside the bundle. In rhodopsin, the Arg of Glu-Arg-Tyr interacts with the adjacent Asp to constrain Arg outside the TMH bundle. V142D was 70% more active than KSHV-GPCR, suggesting that an Arg residue, which is constrained outside the bundle by interacting with Asp-142, leads to a receptor that signals more actively. Because the usually conserved Asp in the middle of TMH2 is not present in KSHV-GPCR, we tested whether Asp-83 at the intracellular end of TMH2 was involved in signaling. D83N and D83A were 110 and 190% more active than KSHV-GPCR, respectively. The double mutant D83A/V142D was 510% more active than KSHV-GPCR. That is, cosubstitutions of Asp-83 by Ala and Val-142 by Asp act synergistically to increase basal signaling. A model of KSHV-GPCR predicts that Arg-143 interacts with residues in the TMH bundle and that the sidechain of Asp-83 does not interact with Arg-143. These data are consistent with the hypothesis that Arg-143 and Asp-83 independently affect the signaling activity of KSHV-GPCR.


Subject(s)
Herpesvirus 8, Human/physiology , Receptors, Chemokine/chemistry , Receptors, Chemokine/physiology , Amino Acid Sequence , Amino Acid Substitution , Base Sequence , Chemokine CXCL12 , Chemokines/pharmacology , Chemokines, CC/pharmacology , Chemokines, CXC/pharmacology , DNA Primers , Herpesvirus 8, Human/genetics , Humans , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Polymerase Chain Reaction , Protein Structure, Secondary , Receptors, Chemokine/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/physiology
10.
J Biol Chem ; 276(10): 7122-8, 2001 Mar 09.
Article in English | MEDLINE | ID: mdl-11116138

ABSTRACT

We coexpressed Kaposi's sarcoma-associated herpesvirus G protein-coupled receptors (KSHV-GPCRs) with thyrotropin-releasing hormone (TRH) receptors or m1-muscarinic-cholinergic receptors in Xenopus oocytes and in mammalian cells. In oocytes, KSHV-GPCR expression resulted in pronounced (81%) inhibition (heterologous desensitization) of Ca(2+)-activated chloride current responses to TRH and acetylcholine. Similar inhibitions of cytoplasmic free Ca(2+) responses to TRH were observed in human embryonic kidney HEK 293 EM cells and in mouse pituitary AtT20 cells. Further study of oocytes showed that this inhibition was partially reversed by interferon-gamma-inducible protein 10 (IP-10), an inverse agonist of KSHV-GPCR. The basal rate of (45)Ca(2+) efflux in oocytes expressing KSHV-GPCRs was 4.4 times greater than in control oocytes, and IP-10 rapidly inhibited increased (45)Ca(2+) efflux. In the absence of IP-10, growth-related oncogene alpha caused a further 2-fold increase in (45)Ca(2+) efflux. In KSHV-GPCR-expressing oocytes, responses to microinjected inositol 1,4,5-trisphosphate were inhibited by 74%, and this effect was partially reversed by interferon-gamma-inducible protein 10. Treatment with thapsigargin suggested that the pool of calcium available for mobilization by TRH was decreased in oocytes coexpressing KSHV-GPCRs. These results suggest that constitutive signaling by KSHV-GPCR causes heterologous desensitization of responses mediated by other receptors, which signal via the phosphoinositide/calcium pathway, which is caused by depletion of intracellular calcium pools.


Subject(s)
Calcium/metabolism , Herpesvirus 8, Human/metabolism , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled , Saccharomyces cerevisiae Proteins , Signal Transduction , Acetylcholine/pharmacology , Animals , Cell Line , Cells, Cultured , Chemokine CXCL10 , Chemokines, CXC/metabolism , Chlorides/metabolism , Cytoplasm/metabolism , Electrophysiology , Enzyme Inhibitors/pharmacology , Humans , Interferon-gamma/metabolism , Mice , Oocytes/metabolism , Receptor, Muscarinic M1 , Receptors, Muscarinic/metabolism , Thapsigargin/pharmacology , Thyrotropin-Releasing Hormone/metabolism , Time Factors , Xenopus
11.
Endocrinology ; 141(10): 3717-22, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11014227

ABSTRACT

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


Subject(s)
Heterotrimeric GTP-Binding Proteins/metabolism , Receptors, Thyrotropin-Releasing Hormone/genetics , Receptors, Thyrotropin-Releasing Hormone/metabolism , Amino Acid Sequence/genetics , Amino Acid Substitution , Animals , COS Cells , Female , GTP-Binding Protein alpha Subunits, Gq-G11 , Ligands , Molecular Conformation , Molecular Sequence Data , Oocytes , Receptors, Thyrotropin-Releasing Hormone/drug effects , Receptors, Thyrotropin-Releasing Hormone/physiology , Signal Transduction/drug effects , Thyrotropin-Releasing Hormone/pharmacology , Xenopus laevis
12.
Neuroendocrinology ; 71(3): 170-6, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10729788

ABSTRACT

Dynorphin (Dyn) peptides were previously shown to increase plasma corticotropin (ACTH) in the ovine fetus, but the site of its action remains unclear. In the present study, Dyn A(1-17) was found to stimulate ACTH release from mouse anterior pituitary tumor AtT-20 cells in a dose-dependent manner. Naloxone did not block the effect of Dyn A(1-17) and the selective kappa-opioid receptor agonist U50488H did not stimulate ACTH release. Dyn A(2-17), a degradative peptide fragment that does not bind to opioid receptors, also stimulated ACTH release from AtT-20 cells. Although the nonopioid effects of Dyn have previously been attributed to N-methyl-D-aspartate (NMDA) receptors, the ACTH-releasing effects of Dyn A(1-17) in AtT-20 cells were not affected by co-administration of NMDA receptor antagonist LY235959. The ACTH response to Dyn A(1-17) could not be blocked by alpha-helical CRH (CRH antagonist) and was additive with a maximal stimulatory dose of CRH, suggesting different mechanisms of action. These results show that the release of ACTH by Dyn A(1-17) in AtT-20 cells is not mediated by kappa-opioid receptors or by the NMDA receptor.


Subject(s)
Adrenocorticotropic Hormone/metabolism , Dynorphins/pharmacology , Narcotics/pharmacology , Peptide Fragments/pharmacology , Pituitary Gland, Anterior/metabolism , Animals , Cell Line , Corticotropin-Releasing Hormone/metabolism , Dynorphins/metabolism , Endorphins/physiology , Excitatory Amino Acid Antagonists/pharmacology , Isoquinolines/pharmacology , Mice , N-Methylaspartate/antagonists & inhibitors , N-Methylaspartate/pharmacology , Narcotics/metabolism , Peptide Fragments/metabolism , Pituitary Gland, Anterior/cytology
14.
Mol Endocrinol ; 14(1): 183-93, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10628757

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.


Subject(s)
Brain/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Receptors, G-Protein-Coupled , Receptors, Thyrotropin-Releasing Hormone/metabolism , 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
15.
J Biol Chem ; 274(44): 31327-32, 1999 Oct 29.
Article in English | MEDLINE | ID: mdl-10531332

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) contains a gene encoding a G protein-coupled receptor (KSHV-GPCR) that is homologous to mammalian chemokine receptors. KSHV-GPCR signals constitutively (in an agonist-independent manner) via the phosphoinositide-inositol 1,4,5-trisphosphate pathway. Because it has been proposed that the N terminus (N-TERM) of other GPCRs may act as tethered agonists, we determined whether the N-TERM of KSHV-GPCR is necessary for constitutive signaling activity or ligand binding, or both. We show that replacement of the entire N-TERM of KSHV-GPCR with those of two other GPCRs, deletion of residues within the N-TERM, and disruption of a putative disulfide bond that may hold the N-TERM in close proximity to extracellular loop 3 do not affect constitutive signaling activity but decrease chemokine binding. There were differences in the effects of mutation of the N-TERM on binding of the chemokines growth-related oncogene alpha, which is an agonist, and interferon-gamma-inducible protein-10, which is an inverse agonist. The effects on chemokine binding were accompanied by changes in chemokine regulation of KSHV-GPCR signaling. We conclude that the N-TERM is not necessary for constitutive KSHV-GPCR signaling, i.e. the N-TERM is not a tethered agonist, but plays a crucial role in binding of chemokine ligands and of chemokine regulation of KSHV-GPCR signaling.


Subject(s)
Chemokines/metabolism , Herpesvirus 8, Human , Intercellular Signaling Peptides and Proteins , Receptors, Chemokine/metabolism , Animals , COS Cells , Chemokine CXCL1 , Chemokine CXCL10 , Chemokines, CXC/metabolism , Chemotactic Factors/metabolism , Growth Substances/metabolism , Inositol Phosphates/metabolism , Ligands , Receptors, CXCR3 , Receptors, Chemokine/genetics , Recombinant Proteins/metabolism , Sequence Deletion , Signal Transduction
16.
Endocrinology ; 140(10): 4916-9, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10499553

ABSTRACT

Two types of rat TRH receptor (TRH-R1 and TRH-R2) have been identified and shown previously to exhibit similar binding and stimulated signaling activity via the phosphoinositide-calcium transduction pathway. Since mouse TRH-R1 exhibits basal (or constitutive or ligand-independent) signaling activity, we compared basal signaling by TRH-R1 and TRH-R2. Basal signaling was measured as receptor-mediated reporter gene induction via different transcription factors. We found that TRH-R2 exhibited higher basal signaling activity than TRH-R1 via pathways mediated by transcription factors AP-1, Elk-1 and CREB. Furthermore, CREB-mediated transcription was directly dependent on the level of TRH-R2 expression and was inhibited by midazolam, a specific inverse agonist of basal TRH-R signaling. Since TRH-R1 and TRH-R2 exhibit distinct anatomic distributions in the rat, it is possible that TRH ligand-independent signaling is more important in tissues/cells in which TRH-R2 is expressed and less important in tissues in which TRH-R1 is found.


Subject(s)
CCAAT-Enhancer-Binding Proteins , Protein Isoforms/physiology , Receptors, Thyrotropin-Releasing Hormone/physiology , Signal Transduction , Animals , Cyclic AMP Response Element-Binding Protein/physiology , DNA-Binding Proteins/physiology , Gene Expression Regulation/physiology , Genes, Reporter/genetics , Humans , Luciferases/genetics , Midazolam/pharmacology , Proto-Oncogene Proteins/physiology , Rats , Receptors, Thyrotropin-Releasing Hormone/agonists , Transcription Factor AP-1/physiology , Transcription Factor CHOP , Transcription Factors/physiology , Transcription, Genetic/drug effects , Transcriptional Activation , ets-Domain Protein Elk-1
17.
Br J Pharmacol ; 126(5): 1097-106, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10204996

ABSTRACT

1. C335Stop is a constitutively active mutant of the TRH receptor (TRH-R). To investigate the mechanism of the decreased responsiveness of C335Stop TRH-R, we studied cellular Ca2+ concentrations ([Ca2+]i) in AtT20 cells stably transfected with C335Stop TRH-R cDNA, or Ca2+-activated chloride currents in Xenopus laevis oocytes expressing this mutant receptor after injection of cRNA. The competitive TRH-R binding antagonist, chlorodiazepoxide (CDE), was used as an inverse agonist to study the contribution of constitutive activity to desensitization. 2. Acute treatment with CDE resulted in a rapid (within minutes) decrease in [Ca2+]i and an increase in the response amplitude to TRH with no measurable change in receptor density. Conversely, removal of chronically administered CDE caused a rapid increase in [Ca2+]i and a decrease in TRH response amplitude. 3. CDE abolished heterologous desensitization induced by C335Stop TRH-R on muscarinic m1-receptor (ml-R) co-expressed in Xenopus oocytes. 4. Chelation of extracellular calcium with EGTA caused a rapid decrease in [Ca2+]i and a concomitant increase in the response to TRH in AtT20 cells expressing C335Stop TRH-Rs. 5. Chelerythrine, a specific inhibitor of protein kinase C (PKC), reversed the heterologous desensitization of the response to acetylcholine (ACh). The phosphoserine/phosphothreonine phosphatase inhibitor, okadaic acid, abolished the effect of chelerythrine. 6. Down-regulation of PKC by chronic exposure to phorbol 12-myristate 13-acetate (PMA) or acute inhibition with chelerythrine caused a partial resensitization of the response to TRH. 7. Western analysis indicated that the alpha subtype of protein kinase C was down-regulated in cells expressing C335Stop TRH-Rs. Following a 5 min exposure to PMA, the residual alphaPKC translocated to the particular fraction. 8. We propose that cells expressing the constitutively active mutant TRH-R rapidly desensitize their response, utilizing a mechanism mediated by an increase in [Ca2+]i and PKC.


Subject(s)
Anti-Anxiety Agents/pharmacology , Calcium/physiology , Protein Kinase C/physiology , Receptors, Thyrotropin-Releasing Hormone/antagonists & inhibitors , Thyrotropin-Releasing Hormone/metabolism , Animals , Benzodiazepines , Mutation , Oocytes/drug effects , Oocytes/metabolism , Receptors, Thyrotropin-Releasing Hormone/genetics , Receptors, Thyrotropin-Releasing Hormone/metabolism , Tumor Cells, Cultured , Xenopus laevis
18.
Biochemistry ; 38(6): 1866-72, 1999 Feb 09.
Article in English | MEDLINE | ID: mdl-10026267

ABSTRACT

Human calcitonin receptor (hCTR) subtypes contain three or four potential Asn-linked glycosylation sites in their extracellular amino termini. The role of glycosylation in hCTR function has not been identified, but it has been suggested that inhibition of glycosylation does not affect binding or signaling. To determine the role of glycosylation in hCTR biology, we studied the effects of inhibition of glycosylation and of substitution of Asn residues that are potential glycosylation sites. Native and mutated hCTRs were studied after transient expression in monkey kidney COS-1 cells. Tunicamycin, administered as part of a treatment protocol that inhibited glycosylation of all expressed receptors, decreased salmon calcitonin (sCT) binding affinities and signaling potencies at hCTRs with three or four potential glycosylation sites. In hCTR3, which contains three potential glycosylation sites at positions 26, 78, and 83, site-specific substitution of Asn-26 by Ala had no effect on sCT binding affinity or potency, whereas substitution of Asn-78 or Asn-83 lowered sCT affinity and potency. A mutant hCTR3 in which all three Asn residues were substituted with Ala exhibited no high-affinity sCT binding and potencies of several calcitonin analogues that were more than 100-fold lower than that of native hCTR3. Our data show that glycosylation is important for high-affinity binding and potency of calcitonin analogues at hCTRs.


Subject(s)
Receptors, Calcitonin/metabolism , Alanine/genetics , Amino Acid Substitution/genetics , Animals , Asparagine/genetics , Binding Sites/genetics , COS Cells , Calcitonin/analogs & derivatives , Calcitonin/metabolism , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Glycosylation/drug effects , Humans , Iodine Radioisotopes , Mutagenesis, Site-Directed , Receptors, Calcitonin/antagonists & inhibitors , Receptors, Calcitonin/genetics , Salmon , Transfection , Tunicamycin/pharmacology
19.
Mol Pharmacol ; 54(6): 968-78, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9855624

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.


Subject(s)
Cell Membrane/metabolism , Receptors, Thyrotropin-Releasing Hormone/chemistry , Tryptophan/chemistry , 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
20.
J Clin Invest ; 102(8): 1469-72, 1998 Oct 15.
Article in English | MEDLINE | ID: mdl-9788958

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV)/human herpesvirus 8, a virus that appears to be involved in the pathogenesis of Kaposi's sarcoma and primary effusion lymphomas, encodes a G protein-coupled receptor (KSHV-GPCR) that exhibits constitutive signaling. In this report, we show that two chemokines, interleukin 8 (IL-8) and growth-related protein-alpha, activate KSHV-GPCR over constitutive levels. Moreover, as with human receptors, the integrity of the ELR motif of these chemokines is required for activation of KSHV-GPCR. Other residues that are required for IL-8 binding to human chemokine receptors CXCR1 and CXCR2 are important for KSHV-GPCR activation also. Thus, it appears that the ELR binding site and other key domains of ELR chemokine activation have been preserved in the virus KSHV-GPCR. The results suggest that KSHV-GPCR originated from CXCR1 or CXCR2 and that activation of KSHV-GPCR by endogenous chemokines may affect the pathobiology of KSHV infection in humans.


Subject(s)
Chemokines/pharmacology , Herpesvirus 8, Human , Intercellular Signaling Peptides and Proteins , Receptors, Chemokine/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Binding Sites , COS Cells , Chemokine CXCL1 , Chemokine CXCL10 , Chemokines, CXC/pharmacology , Chemotactic Factors/pharmacology , Dose-Response Relationship, Drug , GTP-Binding Proteins/metabolism , Growth Substances/pharmacology , Interleukin-8/pharmacology , Mice , Molecular Sequence Data , Oligopeptides/pharmacology , Platelet Factor 4/pharmacology , Protein Binding , Receptors, Chemokine/genetics , Recombinant Proteins/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...