Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
1.
Endocrinology ; 154(8): 2948-58, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23736295

ABSTRACT

Several liganded nuclear receptors have alternative ligands acting in a tissue-specific fashion and playing important biological roles. We present evidence that 3,5-diiodothyronine (T(2)), a naturally occurring iodothyronine that results from T(3) outer-ring deiodination, is an alternative ligand for thyroid hormone receptor ß1 (TRß1). In tilapia, 2 TRß isoforms differing by 9 amino acids in the ligand-binding domain were cloned. Binding and transactivation studies showed that T(2) activates the human and the long tilapia TRß1 isoform, but not the short one. A chimeric human TRß1 (hTRß1) that contained the 9-amino-acid insert showed no response to T(2), suggesting that the conformation of the hTRß1 naturally allows T(2) binding and that other regions of the receptor are implicated in TR activation by T(2). Indeed, further analysis showed that the N terminus is essential for T(2)-mediated transactivation but not for that by T(3) in the long and hTRß1, suggesting a functional interaction between the N-terminal domain and the insertion in the ligand-binding domain. To establish the functional relevance of T(2)-mediated TRß1 binding and activation, mRNA expression and its regulation by T(2) and T(3) was evaluated for both isoforms. Our data show that long TRß1expression is 10(6)-fold higher than that of the short isoform, and T(3) and T(2) differentially regulate the expression of these 2 TRß1 isoforms in vivo. Taken together, our results prompted a reevaluation of the role and mechanism of action of thyroid hormone metabolites previously believed to be inactive. More generally, we propose that classical liganded receptors are only partially locked to very specific ligands and that alternative ligands may play a role in the tissue-specific action of receptors.


Subject(s)
Diiodothyronines/metabolism , Fish Proteins/metabolism , Recombinant Fusion Proteins/metabolism , Thyroid Hormone Receptors beta/metabolism , Animals , Binding Sites/genetics , Binding, Competitive , Cell Line , Cell Line, Tumor , Diiodothyronines/pharmacology , Dose-Response Relationship, Drug , Fish Proteins/agonists , Fish Proteins/genetics , Gene Expression/drug effects , HEK293 Cells , Humans , Kinetics , Ligands , Protein Isoforms/agonists , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Thyroid Hormone Receptors beta/agonists , Thyroid Hormone Receptors beta/genetics , Tilapia , Transcriptional Activation/drug effects , Transfection , Triiodothyronine/metabolism , Triiodothyronine/pharmacology
2.
J Pharm Pharmacol ; 65(4): 528-38, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23488781

ABSTRACT

OBJECTIVES: Semagacestat, is a γ-secretase inhibitor, which belongs to a class of drugs that are being developed as therapeutic agents for Alzheimer's disease (AD). This study aims to evaluate another potential effect of semagacestat, namely its ability to stimulate the growth hormone secretagogue receptor (GHS-R1a), which may also contribute to its therapeutic efficacy. METHODS: The GHS-R1a-activating potential of semagacestat and its synthetic precursor was assessed in an in vitro calcium mobilization assay in cells expressing the GHS-R1a receptor and compared with that of the endogenous peptide GHS-R1a agonist, acyl-ghrelin, as well as the non-peptidyl synthetic GHS-R1a agonist, MK0677. In addition, semagacestat-mediated cellular trafficking of the GHS-R1a receptor, expressed as an enhanced green fluorescent protein tagged fusion protein, was analysed. KEY FINDINGS: Semagacestat and its precursor were shown to activate the GHS-R1a receptor, as demonstrated by an increased GHS-R1a-mediated intracellular calcium influx. Moreover, a synergistic GHS-R1a receptor activation was shown following a combined exposure to ghrelin and semagacestat. In addition, GHS-R1a receptor internalization was observed upon exposure to semagacestat and its precursor. CONCLUSION: These data suggest a novel molecular mechanism of action for semagacestat via modest GHS-R1a receptor activation. Studies focusing on the relative functional consequence of such effects in vivo are now warranted.


Subject(s)
Alanine/analogs & derivatives , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Azepines/pharmacology , Enzyme Inhibitors/pharmacology , Nerve Tissue Proteins/agonists , Neurons/drug effects , Neuroprotective Agents/pharmacology , Receptors, Ghrelin/agonists , Acylation , Alanine/chemical synthesis , Alanine/metabolism , Alanine/pharmacology , Azepines/chemical synthesis , Azepines/metabolism , Calcium Signaling/drug effects , Cell Line, Tumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Ghrelin/analogs & derivatives , Ghrelin/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Indoles/pharmacology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/metabolism , Nootropic Agents/chemical synthesis , Nootropic Agents/metabolism , Nootropic Agents/pharmacology , Prodrugs/chemical synthesis , Prodrugs/metabolism , Prodrugs/pharmacology , Protein Transport/drug effects , Receptors, Ghrelin/genetics , Receptors, Ghrelin/metabolism , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/metabolism , Spiro Compounds/pharmacology , Structure-Activity Relationship
3.
Neuropharmacology ; 64: 45-52, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22735771

ABSTRACT

Positive allosteric modulators of α-amino-3-hydroxy-5-methyl-isoxazole-propionic acid (AMPA) receptors facilitate synaptic plasticity and can improve various forms of learning and memory. These modulators show promise as therapeutic agents for the treatment of neurological disorders such as schizophrenia, ADHD, and mental depression. Three classes of positive modulator, the benzamides, the thiadiazides, and the biarylsulfonamides differentially occupy a solvent accessible binding pocket at the interface between the two subunits that form the AMPA receptor ligand-binding pocket. Here, we describe the electrophysiological properties of a new chemotype derived from a structure-based drug design strategy (SBDD), which makes similar receptor interactions compared to previously reported classes of modulator. This pyrazole amide derivative, JAMI1001A, with a promising developability profile, efficaciously modulates AMPA receptor deactivation and desensitization of both flip and flop receptor isoforms. This article is part of a Special Issue entitled 'Cognitive Enhancers'.


Subject(s)
Acetamides/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Nootropic Agents/pharmacology , Pyrazoles/pharmacology , Receptors, AMPA/agonists , Thiophenes/pharmacology , Acetamides/chemistry , Acetamides/metabolism , Allosteric Site , Benzamides/chemistry , Benzamides/metabolism , Benzamides/pharmacology , Chemistry, Pharmaceutical/methods , Databases, Protein , Drug Design , Excitatory Amino Acid Agonists/chemistry , Excitatory Amino Acid Agonists/metabolism , HEK293 Cells , Humans , Kidney/drug effects , Kidney/metabolism , Kinetics , Ligands , Nootropic Agents/chemistry , Nootropic Agents/metabolism , Protein Conformation , Protein Subunits/agonists , Protein Subunits/chemistry , Protein Subunits/metabolism , Pyrazoles/chemistry , Pyrazoles/metabolism , Receptors, AMPA/chemistry , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/metabolism , Sulfonamides/pharmacology , Thiophenes/chemistry , Thiophenes/metabolism
4.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 28(2): 194-6, 2012 Feb.
Article in Chinese | MEDLINE | ID: mdl-23162922

ABSTRACT

AIM: To establish a drug screening model of CB(2) agonist in vitro based on signal regulation pathway. METHODS: Plasmid pIRES(2)-EGFP-CB(2), pGL(4), 29[luc2P/CRE/Hygro] and PRL-TK were co-transfected into CHO cells in 96 wells plate, to screen agonists of CB(2) receptor by detecting the expressing levels of dual luciferase activity. The concentration and acting time of the agonist were optimized and the stability of the model were investigated. RESULTS: The largest relative induction activity was obtained after 8h drug administration. Establishment of a high throughput screening model for CB(2) receptor agonist. The Z' factor is 0.75 demonstrating its perfect stability. CONCLUSION: Successfully establish a drug screening model of CB(2) agonist, which provided a basis for searching valid material from traditional Chinese medicine.


Subject(s)
Cannabinoid Receptor Agonists/pharmacology , High-Throughput Screening Assays , Receptor, Cannabinoid, CB2/agonists , Animals , CHO Cells , Cricetinae , Dose-Response Relationship, Drug , Receptor, Cannabinoid, CB2/genetics , Receptor, Cannabinoid, CB2/metabolism , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reproducibility of Results
5.
Biotechnol Prog ; 28(6): 1588-97, 2012.
Article in English | MEDLINE | ID: mdl-22961781

ABSTRACT

To avoid high systemic doses, strategies involving antigen-specific delivery of cytokine via linked antibodies or antibody fragments have been used. Targeting cancer-associated peptides presented by major histocompatibility complex (MHC) molecules (pepMHC) increases the number of potential target antigens and takes advantage of cross-presentation on tumor stroma and in draining lymph nodes. Here, we use a soluble, high-affinity single-chain T cell receptor Vα-Vß (scTv), to deliver cytokines to intracellular tumor-associated antigens presented as pepMHC. As typical wild-type T cell receptors (TCRs) exhibit low affinity (K(d) = 1-100 µM or more), we used an engineered TCR, m33, that binds its antigenic peptide SIYRYYGL (SIY) bound to the murine class I major histocompatability complex protein H2-K(b) (SIY/K(b) ) with nanomolar affinity (K(d) = 30 nM). We generated constructs consisting of m33 scTv fused to murine interleukin 2 (IL-2), interleukin 15 (IL-15), or IL-15/IL-15Rα (IL-15 linked to IL-15Rα sushi domain, called "superfusion"). The fusions were purified with good yields and bound specifically to SIY/K(b) with high affinity. Proper cytokine folding and binding were confirmed, and the fusions were capable of stimulating proliferation of cytokine-dependent cells, both when added directly and when presented in trans, bound to cells with the target pepMHC. The m33 superfusion was particularly potent and stable and represents a promising design for targeted antitumor immunomodulation.


Subject(s)
Interleukin-15 Receptor alpha Subunit/chemistry , Interleukin-15/chemistry , Protein Engineering/methods , Receptors, Antigen, T-Cell/chemistry , Recombinant Fusion Proteins/chemistry , Animals , Cell Line, Tumor , Cell Proliferation , Drug Delivery Systems , Humans , Interleukin-15/agonists , Interleukin-15/genetics , Interleukin-15/metabolism , Interleukin-15 Receptor alpha Subunit/agonists , Interleukin-15 Receptor alpha Subunit/genetics , Interleukin-15 Receptor alpha Subunit/metabolism , Mice , Oligopeptides/chemistry , Oligopeptides/metabolism , Protein Binding , Rats , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism
6.
Neuropharmacology ; 63(4): 538-46, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22580377

ABSTRACT

Drug development for nicotinic acetylcholine receptors (nAChR) is challenged by subtype diversity arising from variations in subunit composition. On-target activity for neuronal heteromeric receptors is typically associated with CNS receptors that contain α4 and other subunits, while off-target activity could be associated with ganglionic-type receptors containing α3ß4 binding sites and other subunits, including ß4, ß2, α5, or α3 as a structural subunit in the pentamer. Additional interest in α3 ß4 α5-containing receptors arises from genome-wide association studies linking these genes, and a single nucleotide polymorphism (SNP) in α5 in particular, to lung cancer and heavy smoking. While α3 and ß4 readily form receptors in expression system such as the Xenopus oocyte, since α5 is not required for function, simple co-expression approaches may under-represent α5-containing receptors. We used a concatamer of human α3 and ß4 subunits to form ligand-binding domains, and show that we can force the insertions of alternative structural subunits into the functional pentamers. These α3ß4 variants differ in sensitivity to ACh, nicotine, varenicline, and cytisine. Our data indicated lower efficacy for varenicline and cytisine than expected for ß4-containing receptors, based on previous studies of rodent receptors. We confirm that these therapeutically important α4 receptor partial agonists may present different autonomic-based side-effect profiles in humans than will be seen in rodent models, with varenicline being more potent for human than rat receptors and cytisine less potent. Our initial characterizations failed to find functional effects of the α5 SNP. However, our data validate this approach for further investigations.


Subject(s)
Acetylcholine/metabolism , Ganglia/metabolism , Nerve Tissue Proteins/metabolism , Nicotinic Agonists/metabolism , Nicotinic Antagonists/metabolism , Protein Subunits/metabolism , Receptors, Nicotinic/metabolism , Acetylcholine/agonists , Acetylcholine/antagonists & inhibitors , Alkaloids/metabolism , Alkaloids/pharmacology , Animals , Azocines/metabolism , Azocines/pharmacology , Benzazepines/metabolism , Benzazepines/pharmacology , Drug Partial Agonism , Evoked Potentials/drug effects , Humans , Ligands , Mutagenesis, Site-Directed , Mutant Proteins/agonists , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/genetics , Mutant Proteins/metabolism , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nicotine/agonists , Nicotine/antagonists & inhibitors , Nicotine/metabolism , Nicotinic Agonists/pharmacology , Nicotinic Antagonists/pharmacology , Oocytes/drug effects , Oocytes/metabolism , Protein Subunits/agonists , Protein Subunits/antagonists & inhibitors , Protein Subunits/genetics , Quinolizines/metabolism , Quinolizines/pharmacology , Quinoxalines/metabolism , Quinoxalines/pharmacology , Rats , Receptors, Nicotinic/chemistry , Receptors, Nicotinic/genetics , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/metabolism , Species Specificity , Varenicline , Xenopus laevis
7.
Bioorg Med Chem Lett ; 22(12): 4148-52, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22572579

ABSTRACT

The optimization of a series of thieno[3,2-b]thiophene-2-carboxylic acid derivatives for agonist activity against the GPR35 is reported. Compounds were optimized to achieve ß-arrestin-biased agonism for developing probe molecules that may be useful for elucidating the biology and physiology of GPR35. Compound 13 was identified to the most potent GPR35 agonist, and compounds 30 and 36 exhibited the highest efficacy to cause ß-arrestin translocation.


Subject(s)
Anti-Inflammatory Agents/chemical synthesis , Arrestins/agonists , Receptors, G-Protein-Coupled/agonists , Thiophenes/chemical synthesis , Anti-Inflammatory Agents/pharmacology , Arrestins/chemistry , Arrestins/genetics , Biological Assay , Dose-Response Relationship, Drug , HT29 Cells , Humans , Ligands , Protein Transport/drug effects , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Signal Transduction/drug effects , Structure-Activity Relationship , Thiophenes/pharmacology , beta-Arrestins
8.
Molecules ; 17(3): 2833-54, 2012 Mar 07.
Article in English | MEDLINE | ID: mdl-22399138

ABSTRACT

It is known that the ATP-binding cassette transporter A1 (ABCA1) plays a major role in cholesterol homeostasis and high density lipoprotein (HDL) metabolism. Several laboratories have demonstrated that ABCA1 binding to lipid-poor apolipoprotein A-I (apoA-I) will mediate the assembly of nascent HDL and cellular cholesterol efflux, which suggests a possible receptor-ligand interaction between ABCA1 and apoA-I. In this study, a cell-based-ELISA-like high-throughput screening (HTS) method was developed to identify the synthetic and natural compounds that can regulate binding activity of ABCA1 to apoA-I. The cell-based-ELISA-like high-throughput screen was conducted in a 96-well format using Chinese hamster ovary (CHO) cells stably transfected with ABCA1 pIRE2-EGFP (Enhanced Green Fluorecence Protein) expression vector and the known ABCA1 inhibitor glibenclamide as the antagonist control. From 2,600 compounds, a xanthone compound (IMB 2026791) was selected using this HTS assay, and it was proved as an apoA-I binding agonist to ABCA1 by a flow cytometry assay and western blot analysis. The [3H] cholesterol efflux assay of IMB2026791 treated ABCA1-CHO cells and PMA induced THP-1 macrophages (human acute monocytic leukemia cell) further confirmed the compound as an accelerator of cholesterol efflux in a dose-dependent manner with an EC(50) of 25.23 µM.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Apolipoprotein A-I/metabolism , Cholesterol/metabolism , Xanthones/pharmacology , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/agonists , ATP-Binding Cassette Transporters/antagonists & inhibitors , Animals , CHO Cells , Cell Survival/drug effects , Cricetinae , Dose-Response Relationship, Drug , Glyburide/pharmacology , Green Fluorescent Proteins/metabolism , High-Throughput Screening Assays , Humans , Inhibitory Concentration 50 , Protein Binding , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/metabolism , Tetradecanoylphorbol Acetate/pharmacology
9.
J Med Chem ; 55(5): 2125-43, 2012 Mar 08.
Article in English | MEDLINE | ID: mdl-22329602

ABSTRACT

Two fluorescent derivatives of the M1 muscarinic selective agonist AC-42 were synthesized by coupling the lissamine rhodamine B fluorophore (in ortho and para positions) to AC42-NH(2). This precursor, prepared according to an original seven-step procedure, was included in the study together with the LRB fluorophore (alone or linked to an alkyl chain). All these compounds are antagonists, but examination of their ability to inhibit or modulate orthosteric [(3)H]NMS binding revealed that para-LRB-AC42 shared several properties with AC-42. Carefully designed experiments allowed para-LRB-AC42 to be used as a FRET tracer on EGFP-fused M1 receptors. Under equilibrium binding conditions, orthosteric ligands, AC-42, and the allosteric modulator gallamine behaved as competitors of para-LRB-AC42 binding whereas other allosteric compounds such as WIN 51,708 and N-desmethylclozapine were noncompetitive inhibitors. Finally, molecular modeling studies focused on putative orthosteric/allosteric bitopic poses for AC-42 and para-LRB-AC42 in a 3D model of the human M1 receptor.


Subject(s)
Fluorescent Dyes/chemical synthesis , Molecular Probes/chemical synthesis , Piperidines/chemical synthesis , Receptor, Muscarinic M1/agonists , Receptor, Muscarinic M1/metabolism , Rhodamines/chemical synthesis , Allosteric Regulation , Calcium/metabolism , Fluorescence Resonance Energy Transfer , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacology , Green Fluorescent Proteins/genetics , HEK293 Cells , Humans , Models, Molecular , Molecular Probes/chemistry , Molecular Probes/pharmacology , Piperidines/chemistry , Piperidines/pharmacology , Radioligand Assay , Receptor, Muscarinic M1/antagonists & inhibitors , Receptor, Muscarinic M1/genetics , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Rhodamines/chemistry , Rhodamines/pharmacology , Solubility , Structure-Activity Relationship
10.
Mol Endocrinol ; 26(3): 502-20, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22301784

ABSTRACT

Vasopressin (AVP) and CRH synergistically regulate adrenocorticotropin and insulin release at the level of the pituitary and pancreas, respectively. Here, we first extended these AVP and CRH coregulation processes to the adrenal medulla. We demonstrate that costimulation of chromaffin cells by AVP and CRH simultaneously induces a catecholamine secretion exceeding the one induced by each hormone alone, thus demonstrating a net potentiation. To further elucidate the molecular mechanisms underlying this synergism, we coexpressed human V1b and CRH receptor (CRHR)1 receptor in HEK293 cells. In this heterologous system, AVP also potentiated CRH-stimulated cAMP accumulation in a dose-dependent and saturable manner. This effect was only partially mimicked by phorbol ester or inhibited by a phospholipase C inhibitor respectively. This finding suggests the existence of an new molecular mechanism, independent from second messenger cross talk. Similarly, CRH potentiated the AVP-induced inositol phosphates production. Using bioluminescence resonance energy transfer, coimmunoprecipitation, and receptor rescue experiments, we demonstrate that V1b and CRHR1 receptors assemble as heterodimers. Moreover, new pharmacological properties emerged upon receptors cotransfection. Taken together, these data strongly suggest that direct molecular interactions between V1b and CRHR1 receptors play an important role in mediating the synergistic interactions between these two receptors.


Subject(s)
Corticotropin-Releasing Hormone/physiology , Protein Multimerization , Receptors, Corticotropin-Releasing Hormone/metabolism , Receptors, Vasopressin/metabolism , Vasopressins/physiology , Adrenal Glands/cytology , Animals , Antidiuretic Hormone Receptor Antagonists , Catecholamines/metabolism , Cattle , Cells, Cultured , Chromaffin Cells/metabolism , Corticotropin-Releasing Hormone/pharmacology , Cyclic AMP/metabolism , Enzyme Activation , Estrenes/pharmacology , HEK293 Cells , Humans , Hydrocarbons, Halogenated/pharmacology , Indoles/pharmacology , Inositol Polyphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/metabolism , Primary Cell Culture , Protein Binding , Pyrrolidines/pharmacology , Pyrrolidinones/pharmacology , Receptor Cross-Talk , Receptors, Corticotropin-Releasing Hormone/agonists , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Vasopressin/agonists , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/metabolism , Second Messenger Systems , Thiazines/pharmacology , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/metabolism , Vasopressins/pharmacology
11.
J Biol Chem ; 287(8): 5301-9, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22215671

ABSTRACT

Numerous receptors for ATP, ADP, and adenosine exist; however, it is currently unknown whether a receptor for the related nucleotide adenosine 5'-monophosphate (AMP) exists. Using a novel cell-based assay to visualize adenosine receptor activation in real time, we found that AMP and a non-hydrolyzable AMP analog (deoxyadenosine 5'-monophosphonate, ACP) directly activated the adenosine A(1) receptor (A(1)R). In contrast, AMP only activated the adenosine A(2B) receptor (A(2B)R) after hydrolysis to adenosine by ecto-5'-nucleotidase (NT5E, CD73) or prostatic acid phosphatase (PAP, ACPP). Adenosine and AMP were equipotent human A(1)R agonists in our real-time assay and in a cAMP accumulation assay. ACP also depressed cAMP levels in mouse cortical neurons through activation of endogenous A(1)R. Non-selective purinergic receptor antagonists (pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid and suramin) did not block adenosine- or AMP-evoked activation. Moreover, mutation of His-251 in the human A(1)R ligand binding pocket reduced AMP potency without affecting adenosine potency. In contrast, mutation of a different binding pocket residue (His-278) eliminated responses to AMP and to adenosine. Taken together, our study indicates that the physiologically relevant nucleotide AMP is a full agonist of A(1)R. In addition, our study suggests that some of the physiological effects of AMP may be direct, and not indirect through ectonucleotidases that hydrolyze this nucleotide to adenosine.


Subject(s)
Adenosine A1 Receptor Agonists/pharmacology , Adenosine Monophosphate/pharmacology , Receptor, Adenosine A1/metabolism , 5'-Nucleotidase/metabolism , Adenosine/metabolism , Adenosine A1 Receptor Agonists/chemistry , Adenosine A1 Receptor Agonists/metabolism , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/metabolism , Animals , Cerebral Cortex/cytology , Colforsin/pharmacology , HEK293 Cells , Histidine , Humans , Hydrolysis/drug effects , Ligands , Mice , Molecular Imaging , Neurons/drug effects , Neurons/metabolism , Receptor, Adenosine A1/chemistry , Receptor, Adenosine A2B/metabolism , Receptors, Purinergic P2Y/metabolism , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Single-Cell Analysis
12.
J Med Chem ; 55(3): 1147-60, 2012 Feb 09.
Article in English | MEDLINE | ID: mdl-22216892

ABSTRACT

Bivalent histamine H(2) receptor (H(2)R) agonists were synthesized by connecting pharmacophoric 3-(2-amino-4-methylthiazol-5-yl)-, 3-(2-aminothiazol-5-yl)-, 3-(imidazol-4-yl)-, or 3-(1,2,4-triazol-5-yl)propylguanidine moieties by N(G)-acylation with alkanedioic acids of various chain lengths. The compounds were investigated for H(2)R agonism in GTPase and [(35)S]GTPγS binding assays at guinea pig (gp) and human (h) H(2)R-Gsα(S) fusion proteins including various H(2)R mutants, at the isolated gp right atrium, and in GTPase assays for activity on recombinant H(1), H(3), and H(4) receptors. The bivalent ligands are H(2)R partial or full agonists, up to 2 orders of magnitude more potent than monovalent acylguanidines and, with octanedioyl or decanedioyl spacers, up to 4000 times more potent than histamine at the gpH(2)R. In contrast to their imidazole analogues, the aminothiazoles are highly selective for H(2)R vs other HR subtypes. Compounds with (theoretically) sufficient spacer length (20 CH(2) groups) to simultaneously occupy two orthosteric binding sites in H(2)R dimers are nearly inactive, whereas the highest potency resides in compounds with considerably shorter spacers. Thus, there is no evidence for interaction with H(2)R dimers. The high agonistic potency may result from interaction with an accessory binding site at the same receptor protomer.


Subject(s)
Guanidines/chemical synthesis , Histamine Agonists/chemical synthesis , Receptors, Histamine H2/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , GTP-Binding Protein alpha Subunits, Gs/genetics , Guanidines/chemistry , Guanidines/pharmacology , Guinea Pigs , Heart/drug effects , Heart/physiology , Histamine Agonists/chemistry , Histamine Agonists/pharmacology , Humans , In Vitro Techniques , Ligands , Mutation , Promoter Regions, Genetic , Protein Multimerization , Receptors, Histamine H2/genetics , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/genetics , Structure-Activity Relationship
13.
Exp Hematol ; 40(5): 386-92, 2012 May.
Article in English | MEDLINE | ID: mdl-22269115

ABSTRACT

Preclinical in vivo assessment of the pharmacologic activity of nonpeptidyl thrombopoietin receptor (TPOR) agonists is very difficult because of the high species specificity of such agonists. In this study, we have developed a novel and simple in vivo hollow-fiber assay to preclinically evaluate TPOR agonists. The 32D-mpl cell line was generated by stable transfection of human TPOR into 32D lymphoblast cells and shown to be a specific model for nonpeptide TPOR agonists in vitro. Stably transfected 32D-mpl cells were then sealed in hollow fibers and implanted into nude mice. Cells in hollow fibers specifically responded to TPOR agonists, including thrombopoietin and eltrombopag, a nonpeptide small-molecule TPOR agonist, but not to granulocyte colony-stimulating factor or erythropoietin. Oral administration of eltrombopag stimulated 32D-mpl cell proliferation, prevented 32D-mpl cell apoptosis, and stimulated the phosphorylation of cellular signaling transducers and activators of transcription in a TPOR- and dose-dependent manner. These results indicate that the hollow-fiber assay is a specific and efficient model for rapidly evaluating the in vivo activity of small-molecule TPOR agonists.


Subject(s)
Drug Evaluation, Preclinical/instrumentation , Receptors, Thrombopoietin/agonists , Animals , Benzoates/pharmacology , Cell Line/transplantation , Colony-Forming Units Assay , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Erythropoietin/pharmacology , Female , Granulocyte Colony-Stimulating Factor/pharmacology , Humans , Hydrazines/pharmacology , Implants, Experimental , Interleukin-3/pharmacology , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Pyrazoles/pharmacology , Recombinant Fusion Proteins/agonists , Recombinant Proteins/pharmacology , STAT3 Transcription Factor/metabolism , Thrombopoietin/pharmacology , Transfection
14.
Cell Signal ; 24(1): 106-18, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21893192

ABSTRACT

We previously showed that phorbol-12-myristate-13-acetate (PMA) mediates a robust PKC-dependent sensitization and desensitization of the highly homologous human Gs protein and adenylyl cyclase (AC)-linked D1 (hD1R) and D5 (hD5R) dopaminergic receptors, respectively. Here, we demonstrate using forskolin-mediated AC stimulation that PMA-mediated hD1R sensitization and hD5R desensitization is not associated with changes in AC activity. We next employed a series of chimeric hD1R and hD5R to delineate the underlying structural determinants dictating the subtype-specific regulation of human D1-like receptors by PMA. We first used chimeric receptors in which the whole terminal region (TR) spanning from the extracellular face of transmembrane domain 6 to the end of cytoplasmic tail (CT) or CT alone were exchanged between hD1R and hD5R. CT and TR swaps lead to chimeric hD1R and hD5R retaining PMA-induced sensitization and desensitization of wild type parent receptors. In striking contrast, hD1R sensitization and hD5R desensitization mediated by PMA are correspondingly switched to PMA-induced receptor desensitization and sensitization following the IL3 swap between hD1R and hD5R. Cell treatment with the PKC blocker, Gö6983, inhibits PMA-induced regulation of these chimeric receptors in a similar fashion to wild type receptors. Further studies with chimeras constructed by exchanging IL3 and TR show that PMA-induced regulation of these chimeras remains fully switched relative to their respective wild type parent receptor. Interestingly, results obtained with the exchange of IL3 and TR also reveal that the D1-like subtype-specific regulation by PMA, while fully dictated by IL3, can be modulated in a receptor conformation-dependent manner. Overall, our results strongly suggest that IL3 is the critical determinant underlying the subtype-specific regulation of human D1-like receptor responsiveness by PKC.


Subject(s)
Protein Kinase C/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D5/metabolism , Recombinant Fusion Proteins/metabolism , Adenylyl Cyclases/metabolism , Amino Acid Motifs , Colforsin/pharmacology , Cyclic AMP/metabolism , Dopamine/pharmacology , Dopamine/physiology , Dose-Response Relationship, Drug , Enzyme Activators/pharmacology , HEK293 Cells , Humans , Indoles/pharmacology , Isoenzymes/metabolism , Maleimides/pharmacology , Phosphorylation , Protein Conformation , Protein Kinase C/antagonists & inhibitors , Receptors, Dopamine D1/agonists , Receptors, Dopamine D1/chemistry , Receptors, Dopamine D5/agonists , Receptors, Dopamine D5/chemistry , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/chemistry , Tetradecanoylphorbol Acetate/pharmacology
15.
FEBS J ; 279(2): 211-22, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22059479

ABSTRACT

IκB-ζ, an essential inflammatory regulator, is specifically induced by Toll-like receptor ligands or interleukin (IL)-1ß by post-transcriptional activation mediated via a 165-nucleotide element in IκB-ζ mRNA. Here, we analyzed the Toll-like receptor-IL-1 receptor signaling components involved in the post-transcriptional regulation of IκB-ζ with mutated estrogen receptor [ER(T2)] fusion proteins. Upon 4-hydroxytamoxifen treatment, the ER(T2) fusion proteins with IL-1 receptor-associated kinase (IRAK)1 and IRAK4 elicited specific activation of a reporter gene for the post-transcriptional regulation of IκB-ζ. The tumor necrosis factor receptor-associated factor (TRAF)6-ER(T2) protein activated nuclear factor-κB, but not post-transcriptional regulation, indicating that activation of IRAK1/4, but not of TRAF6, is sufficient to activate the 165-nucleotide element-mediated post-transcriptional mechanism. Interestingly, the post-transcriptional mechanism was not activated in TRAF6-deficient cells, indicating an essential role for TRAF6. Thus, the signaling pathway leading to nuclear factor-κB activation and the post-transcriptional activation bifurcates at IRAK1, suggesting a new pathway activated by IRAK1.


Subject(s)
Interleukin-1 Receptor-Associated Kinases/metabolism , Nuclear Proteins/metabolism , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , Adaptor Proteins, Signal Transducing , Animals , Cells, Cultured , Embryo, Mammalian/cytology , Estrogen Receptor Modulators/pharmacology , Estrogen Receptor beta/agonists , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Gene Silencing , Genes, Reporter/drug effects , HEK293 Cells , Humans , I-kappa B Proteins , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Interleukin-1 Receptor-Associated Kinases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Nuclear Proteins/genetics , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/metabolism , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism
16.
Toxicol Lett ; 206(2): 158-65, 2011 Oct 10.
Article in English | MEDLINE | ID: mdl-21782008

ABSTRACT

Traditional risk assessment of potential endocrine-disruptive pollutants, including PCBs, focus mainly on the effects of parent compounds. Still, biotransformation results in systemic exposure to PCBs and their bioactive metabolites. In the present paper, the effects of twenty ultra-pure non-dioxin-like (NDL) PCBs and their environmentally relevant hydroxy- (OH-) and methylsulfonyl- (MeSO(2)-) metabolites on aromatase activity and their glucocorticoid properties were investigated. Although most NDL-PCBs were inactive, PCB28 inhibited aromatase activity in human placenta microsomes with an IC(50) of 2.2µM. Most of these NDL-PCBs were weak (ant-)agonist of the glucocorticoid receptor (GR). Interestingly, four OH-metabolites of the commonly found NDL-PCB180 were able to inhibit aromatase activity (LOECs in the low µM range) and showed anti-glucocorticoid properties (LOECs in the low nM range), in a concentration-dependent manner. Further, four MeSO(2)-PCBs slightly inhibited aromatase activity and showed anti-glucocorticoid properties. Although, these effects were also associated with cytotoxicity, they were dependent on the position of the MeSO(2)-group on the biphenyl ring. Our results are the first to show that OH-PCBs are both anti-glucocorticoids and aromatase inhibitors. Taken together, these results for PCBs again support the common idea that risk assessment of the endocrine disruptive potential of PCBs should also include their metabolites.


Subject(s)
Aromatase Inhibitors/toxicity , Endocrine Disruptors/toxicity , Mesylates/toxicity , Polychlorinated Biphenyls/toxicity , Receptors, Glucocorticoid/antagonists & inhibitors , Aromatase/biosynthesis , Aromatase/chemistry , Aromatase Inhibitors/chemistry , Aromatase Inhibitors/metabolism , Cell Line , Cell Survival/drug effects , Endocrine Disruptors/chemistry , Endocrine Disruptors/metabolism , Enzyme Induction/drug effects , Female , Genes, Reporter/drug effects , Humans , Hydroxylation , Mesylates/chemistry , Microsomes/enzymology , Osmolar Concentration , Placenta/enzymology , Polychlorinated Biphenyls/chemistry , Polychlorinated Biphenyls/metabolism , Pregnancy , Pregnancy Proteins/antagonists & inhibitors , Receptors, Glucocorticoid/agonists , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Response Elements/drug effects , Risk Assessment/methods
17.
Chem Biol ; 18(6): 752-65, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21700211

ABSTRACT

The NF-E2-related factor 2 (Nrf2) is a key transcriptional regulator of antioxidant defense and detoxification. To directly monitor stabilization of Nrf2, we fused its Neh2 domain, responsible for the interaction with its nucleocytoplasmic regulator, Keap1, to firefly luciferase (Neh2-luciferase). We show that Neh2 domain is sufficient for recognition, ubiquitination, and proteasomal degradation of Neh2-luciferase fusion protein. The Neh2-luc reporter system allows direct monitoring of the adaptive response to redox stress and classification of drugs based on the time course of reporter activation. The reporter was used to screen the Spectrum library of 2000 biologically active compounds to identify activators of Nrf2. The most robust and yet nontoxic Nrf2 activators found--nordihydroguaiaretic acid, fisetin, and gedunin--induced astrocyte-dependent neuroprotection from oxidative stress via an Nrf2-dependent mechanism.


Subject(s)
Genes, Reporter , High-Throughput Screening Assays , NF-E2-Related Factor 2/metabolism , Binding Sites , Cell Line, Tumor , Computer Simulation , Flavonoids/chemistry , Flavonoids/pharmacology , Flavonols , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Kelch-Like ECH-Associated Protein 1 , Kinetics , Limonins/chemistry , Limonins/pharmacology , Luciferases/genetics , Luciferases/metabolism , Masoprocol/chemistry , Masoprocol/pharmacology , NF-E2-Related Factor 2/agonists , NF-E2-Related Factor 2/genetics , Oxidative Stress , Protein Structure, Tertiary , RNA Interference , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Structure-Activity Relationship , Ubiquitination
18.
J Biol Chem ; 286(26): 23334-44, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21543336

ABSTRACT

Toll-like receptor 4 (TLR4) is involved in activation of the innate immune response in a large number of different diseases. Despite numerous studies, the role of separate domains of TLR4 in the regulation of receptor activation is poorly understood. Replacement of the TLR4 ectodomain with LPS-binding proteins MD-2 or CD14 resulted in a robust ligand-independent constitutive activation comparable with the maximal stimulation of the receptor with LPS. The same effect was achieved by the replacement of the ectodomain with a monomeric fluorescent protein or a 24-kDa gyrase B fragment. This demonstrates an intrinsic dimerization propensity of the transmembrane and cytoplasmic domains of TLR4 and reveals a previously unknown function of the ectodomain in inhibiting spontaneous receptor dimerization. Constitutive activation was abolished by the replacement of the ectodomain by a bulkier protein ovalbumin. N-terminal deletion variants of TLR4 revealed that the smallest segment of the ectodomain that already prevents constitutive activity comprises only 90 residues (542 to 631) of the total 608 residues. We conclude that TLR4 represents a receptor with a low threshold of activation that can be rapidly activated by the release of inhibition exerted by its ectodomain. This is important for the sensitivity of TLR4 to activation by different agonists. The TLR4 ectodomain has multiple roles in enabling ligand regulated activation, providing proper localization while serving as an inhibitor to prevent spontaneous, ligand-independent dimerization.


Subject(s)
Protein Multimerization/physiology , Toll-Like Receptor 4/metabolism , HEK293 Cells , Humans , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/immunology , Lipopolysaccharide Receptors/metabolism , Lymphocyte Antigen 96/agonists , Lymphocyte Antigen 96/genetics , Lymphocyte Antigen 96/immunology , Lymphocyte Antigen 96/metabolism , Protein Structure, Tertiary , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
19.
FEBS Lett ; 585(9): 1368-74, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21510948

ABSTRACT

We wanted to analyze the basis for the distinction between OX(1) and OX(2) orexin receptors by the known agonists, orexin-A, orexin-B and Ala(11), D-Leu(15)-orexin-B, of which the latter two show some selectivity for OX(2). For this, chimaeric OX(1)/OX(2) and OX(2)/OX(1) orexin receptors were generated. The receptors were transiently expressed in HEK-293 cells, and potencies of the agonists to elicit cytosolic Ca(2+) elevation were measured. The results show that the N-terminal regions of the receptor are most important, and the exchange of the area from the C-terminal part of the transmembrane helix 2 to the transmembrane helix 4 is enough to lead to an almost total change of the receptor's ligand profile.


Subject(s)
Intracellular Signaling Peptides and Proteins/pharmacology , Neuropeptides/pharmacology , Receptors, G-Protein-Coupled/agonists , Receptors, Neuropeptide/agonists , Recombinant Fusion Proteins/agonists , Amino Acid Sequence , Binding Sites/genetics , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Models, Molecular , Molecular Sequence Data , Mutation , Neuropeptides/chemistry , Orexin Receptors , Orexins , Protein Isoforms/agonists , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Structure, Secondary , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, Neuropeptide/chemistry , Receptors, Neuropeptide/genetics , Recombinant Fusion Proteins/chemistry
20.
Structure ; 19(1): 37-44, 2011 Jan 12.
Article in English | MEDLINE | ID: mdl-21220114

ABSTRACT

The constitutive androstane receptor (CAR) transactivation can occur in the absence of exogenous ligand and this activity is enhanced by agonists TCPOBOP and meclizine. We use biophysical and cell-based assays to show that increased activity of CAR(TCPOBOP) relative to CAR(meclizine) corresponds to a higher affinity of CAR(TCPOBOP) for the steroid receptor coactivator-1. Additionally, steady-state fluorescence spectra suggest conformational differences between CAR(TCPOBOP):RXR and CAR(meclizine):RXR. Hydrogen/deuterium exchange (HDX) data indicate that the CAR activation function 2 (AF-2) is more stable in CAR(TCPOBOP):RXR and CAR(meclizine):RXR than in CAR:RXR. HDX kinetics also show significant differences between CAR(TCPOBOP):RXR and CAR(meclizine):RXR. Unlike CAR(meclizine):RXR, CAR(TCPOBOP):RXR shows a higher overall stabilization that extends into RXR. We identify residues 339-345 in CAR as an allosteric regulatory site with a greater magnitude reduction in exchange kinetics in CAR(TCPOBOP):RXR than CAR(meclizine):RXR. Accordingly, assays with mutations on CAR at leucine-340 and leucine-343 confirm this region as an important determinant of CAR activity.


Subject(s)
Meclizine/pharmacology , Pyridines/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Recombinant Fusion Proteins/metabolism , Retinoid X Receptors/metabolism , Allosteric Site , Animals , Binding Sites , Constitutive Androstane Receptor , Humans , Mice , Models, Molecular , Nuclear Receptor Coactivator 1/chemistry , Protein Binding , Protein Multimerization , Protein Stability , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/chemistry , Recombinant Fusion Proteins/agonists , Recombinant Fusion Proteins/chemistry , Retinoid X Receptors/chemistry , Thermodynamics , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL