Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Oncogene ; 38(15): 2675-2689, 2019 04.
Article in English | MEDLINE | ID: mdl-30538297

ABSTRACT

Targeting of histone methylation has therapeutic potential in oncology. Here, we provide proof-of-principle that pharmacological inhibition of KDM5 histone-demethylases is a new strategy for the personalized treatment of HER2+ breast cancer. The anti-proliferative effects of the prototype of a new class of selective KDM5-inhibitors (KDM5-inh1) are evaluated in 40 cell lines, recapitulating the heterogeneity of breast cancer. This analysis demonstrates that HER2+ cells are particularly sensitive to KDM5 inhibition. The results are confirmed in an appropriate in vivo model with a close structural analog (KDM5-inh1A). RNA-seq data obtained in HER2+ BT-474 cells exposed to KDM5-Inh1 indicate that the compound alters expression of numerous genes downstream of the ERBB2 gene-product, HER2. In selected HER2-positive breast-cancer cells, we demonstrate synergistic interactions between KDM5-inh1 and HER2-targeting agents (trastuzumab and lapatinib). In addition, HER2+ cell lines with innate and acquired resistance to trastuzumab show sensitivity to KDM5-inh1. The levels of KDM5A/B/C proteins, which are selectively targeted by the agent, have no significant association with KDM5-inh1 responsiveness across our panel of breast-cancer cell lines, suggesting the existence of other determinants of sensitivity. Using RNA-seq data of the breast-cancer cell lines we generate a gene-expression model that is a robust predictor of KDM5-inh1 sensitivity. In a test set of breast cancers, this model predicts sensitivity to the compound in a large fraction of HER2+ tumors. In conclusion, KDM5 inhibition has potential in the treatment of HER2+ breast cancer and our gene-expression model can be developed into a diagnostic tool for the selection of patients.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , Receptor, ErbB-2/genetics , Retinoblastoma-Binding Protein 2/genetics , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Trastuzumab/pharmacology
2.
J Med Chem ; 60(3): 886-898, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28045522

ABSTRACT

The G-protein-coupled receptor 39 (GPR39) is a G-protein-coupled receptor activated by Zn2+. We used a homology model-based approach to identify small-molecule pharmacological tool compounds for the receptor. The method focused on a putative binding site in GPR39 for synthetic ligands and knowledge of ligand binding to other receptors with similar binding pockets to select iterative series of minilibraries. These libraries were cherry-picked from all commercially available synthetic compounds. A total of only 520 compounds were tested in vitro, making this method broadly applicable for tool compound development. The compounds of the initial library were inactive when tested alone, but lead compounds were identified using Zn2+ as an allosteric enhancer. Highly selective, highly potent Zn2+-independent GPR39 agonists were found in subsequent minilibraries. These agonists identified GPR39 as a novel regulator of gastric somatostatin secretion.


Subject(s)
Receptors, G-Protein-Coupled/agonists , Zinc/metabolism , Allosteric Regulation , Drug Discovery , Gastric Mucosa/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Molecular Structure , Receptors, G-Protein-Coupled/metabolism , Structure-Activity Relationship
4.
J Med Chem ; 54(8): 2658-67, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21410292

ABSTRACT

The main disadvantages of peptide pharmaceuticals are their rapid degradation and excretion, their low hydrophilicity, and low shelf lifes. These bottlenecks can be circumvented by acylation with fatty acids (lipidation) or polyethylene glycol (PEGylation). Here, we describe the modification of a human pancreatic polypeptide analogue specific for the human (h)Y(2) and hY(4) receptor with PEGs of different size and palmitic acid. Receptor specificity was demonstrated by competitive binding studies. Modifications had only a small influence on binding affinities and no influence on secondary structure. Both modifications improved pharmacokinetic properties of the hPP analogue in vivo and in vitro, however, lipidation showed a greater resistance to degradation and excretion than PEGylation. Furthermore, the lipidated peptide is taken up and degraded solely by the liver but not the kidneys. Lipidation resulted in prolonged action of the hPP analogue in respect of reducing food intake in mice after subcutaneous administration. Therefore, the lipidated hPP analogue could constitute a potential new therapeutic agent against obesity.


Subject(s)
Lipids/chemistry , Pancreatic Polypeptide/metabolism , Amino Acid Sequence , Animals , Biological Availability , Humans , Hydrolysis , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Pancreatic Polypeptide/blood , Pancreatic Polypeptide/chemistry , Protein Structure, Secondary , Rats , Rats, Wistar , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
5.
J Biol Chem ; 282(37): 27354-27365, 2007 Sep 14.
Article in English | MEDLINE | ID: mdl-17599916

ABSTRACT

AMD3465 is a novel, nonpeptide CXCR4 antagonist and a potent inhibitor of HIV cell entry in that one of the four-nitrogen cyclam rings of the symmetrical, prototype bicyclam antagonist AMD3100 has been replaced by a two-nitrogen N-pyridinylmethylene moiety. This substitution induced an 8-fold higher affinity as determined against (125)I-12G5 monoclonal CXCR4 antibody binding, and a 22-fold higher potency in inhibition of CXCL12-induced signaling through phosphatidylinositol accumulation. Mutational mapping of AMD3465 and a series of analogs of this in a library of 23 mutants covering the main ligand binding pocket of the CXCR4 receptor demonstrated that the single cyclam ring of AMD3465 binds in the pocket around AspIV:20 (Asp(171)), in analogy with AMD3100, whereas the N-pyridinylmethylene moiety mimics the other cyclam ring through interactions with the two acidic anchor-point residues in transmembrane (TM)-VI (AspVI:23/Asp(262)) and TM-VII (GluVII:06/Glu(288)). Importantly, AMD3465 has picked up novel interaction sites, for example, His(281) located at the interface of extracellular loop 3 and TM-VII and HisIII:05 (His(113)) in the middle of the binding pocket. It is concluded that the simple N-pyridinylmethylene moiety of AMD3465 substitutes for one of the complex cyclam moieties of AMD3100 through an improved and in fact expanded interaction pattern mainly with residues located in the extracellular segments of TM-VI and -VII of the CXCR4 receptor. It is suggested that the remaining cyclam ring of AMD3465, which ensures the efficacious blocking of the receptor, in a similar manner can be replaced by chemical moieties allowing for, for example, oral bioavailability.


Subject(s)
Anti-HIV Agents/pharmacology , Heterocyclic Compounds/pharmacology , Pyridines/pharmacology , Receptors, CXCR4/antagonists & inhibitors , Amino Acid Sequence , Animals , Antibodies, Monoclonal/metabolism , Benzylamines , COS Cells , Chlorocebus aethiops , Cyclams , Humans , Molecular Sequence Data , Mutation , Pyridines/metabolism , Receptors, CXCR4/chemistry
6.
Endocrinology ; 148(1): 13-20, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16959833

ABSTRACT

GPR39 is an orphan member of the ghrelin receptor family that recently was suggested to be the receptor for obestatin, a peptide derived from the ghrelin precursor. Here, we compare the effect of obestatin to the effect of Zn(2+) on signal transduction and study the effect of obestatin on food intake. Although Zn(2+) stimulated inositol phosphate turnover, cAMP production, arrestin mobilization, as well as cAMP response element-dependent and serum response element-dependent transcriptional activity in GPR39-expressing cells as opposed to mock-transfected cells, no reproducible effect was obtained with obestatin in the GPR39-expressing cells. Moreover, no specific binding of obestatin could be detected in two different types of GPR39-expressing cells using three different radioiodinated forms of obestatin. By quantitative PCR analysis, GPR39 expression was readily detected in peripheral organs such as duodenum and kidney but not in the pituitary and hypothalamus, i.e. presumed central target organs for obestatin. Obestatin had no significant and reproducible effect on acute food intake in either freely fed or fasted lean mice. It is concluded that GPR39 is probably not the obestatin receptor. In contrast, the potency and efficacy of Zn(2+) in respect of activating signaling indicates that this metal ion could be a physiologically relevant agonist or modulator of GPR39.


Subject(s)
Peptide Hormones/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Zinc/metabolism , Animals , Arrestin/metabolism , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetinae , Cricetulus , Cyclic AMP/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Eating/drug effects , Gene Expression/physiology , Genes, Reporter , Ghrelin , Humans , Inositol Phosphates/metabolism , Integrases/genetics , Kidney/cytology , Luciferases/genetics , Mice , Mice, Inbred C57BL , Peptide Hormones/pharmacology , Polymerase Chain Reaction , Receptors, G-Protein-Coupled/genetics , Signal Transduction/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , Tritium , Zinc/pharmacology
7.
J Med Chem ; 49(23): 6638-41, 2006 Nov 16.
Article in English | MEDLINE | ID: mdl-17154491

ABSTRACT

Hits from an in silico derived focused library for CRTH2 were transformed into highly selective antagonists with favorable ADME properties. Oral administration of 4-bromo-2-(1-phenyl-1H-pyrazole-4-carbonyl)phenoxyacetic acid (19) inhibited peribronchial eosinophilia and mucus cell hyperplasia in a mouse model of allergic asthma, supporting the therapeutic potential of this novel compound class. In addition, this selective pharmacological tool compound provides further evidence for CRTH2 as a relevant therapeutic target for treatment of Th2- and eosinophil-related inflammation.


Subject(s)
Acetates/chemistry , Anti-Allergic Agents/chemistry , Pyrazoles/chemistry , Receptors, Immunologic/antagonists & inhibitors , Receptors, Prostaglandin/antagonists & inhibitors , Acetates/chemical synthesis , Acetates/pharmacology , Animals , Anti-Allergic Agents/chemical synthesis , Anti-Allergic Agents/pharmacology , Asthma/drug therapy , Asthma/immunology , Binding, Competitive , Biological Availability , Eosinophils/immunology , Fluorescence Resonance Energy Transfer , Humans , In Vitro Techniques , Inflammation/drug therapy , Inflammation/immunology , Mice , Models, Molecular , Phenoxyacetates , Pyrazoles/chemical synthesis , Pyrazoles/pharmacology , Radioligand Assay , Rats , Stereoisomerism , Structure-Activity Relationship , Th2 Cells/immunology
8.
Bioorg Med Chem Lett ; 16(13): 3380-2, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16678410

ABSTRACT

We herein report a group of allosteric inhibitors of integrin alpha(2)beta(1) based on an arylamide scaffold. Compound 4 showed an IC(50) of 4.80 microM in disrupting integrin I-domain/collagen binding in an ELISA. These arylamide compounds are able to block collagen binding to integrin alpha(2)beta(1) on the platelet surface. Further we find that compound 4 recognizes a hydrophobic cleft on the side of the alpha(2) I-domain, suggesting an alternative targeting site for drug development.


Subject(s)
Amides/pharmacology , Collagen Type I/antagonists & inhibitors , Integrin alpha2beta1/antagonists & inhibitors , Allosteric Regulation , Amides/chemistry , Binding Sites/drug effects , Collagen Type I/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Hydrophobic and Hydrophilic Interactions , Integrin alpha2beta1/metabolism , Magnetic Resonance Spectroscopy/methods , Models, Molecular , Molecular Structure , Protein Binding/drug effects , Stereoisomerism , Structure-Activity Relationship
9.
J Biol Chem ; 281(25): 17337-17346, 2006 Jun 23.
Article in English | MEDLINE | ID: mdl-16567806

ABSTRACT

Much evidence indicates that, during activation of seven-transmembrane (7TM) receptors, the intracellular segments of the transmembrane helices (TMs) move apart with large amplitude, rigid body movements of especially TM-VI and TM-VII. In this study, AspIII:08 (Asp113), the anchor point for monoamine binding in TM-III, was used as the starting point to engineer activating metal ion sites between the extracellular segments of the beta2-adrenergic receptor. Cu(II) and Zn(II) alone and in complex with aromatic chelators acted as potent (EC50 decreased to 0.5 microm) and efficacious agonists in sites constructed between positions III:08 (Asp or His), VI:16 (preferentially Cys), and/or VII:06 (preferentially Cys). In molecular models built over the backbone conformation of the inactive rhodopsin structure, the heavy atoms that coordinate the metal ion were located too far away from each other to form high affinity metal ion sites in both the bidentate and potential tridentate settings. This indicates that the residues involved in the main ligand-binding pocket will have to move closer to each other during receptor activation. On the basis of the distance constraints from these activating metal ion sites, we propose a global toggle switch mechanism for 7TM receptor activation in which inward movement of the extracellular segments of especially TM-VI and, to some extent, TM-VII is coupled to the well established outward movement of the intracellular segments of these helices. We suggest that the pivots for these vertical seesaw movements are the highly conserved proline bends of the involved helices.


Subject(s)
Cell Membrane/metabolism , Metals/chemistry , Amino Acid Sequence , Animals , COS Cells , Cattle , Chlorocebus aethiops , Copper/chemistry , Molecular Conformation , Molecular Sequence Data , Protein Binding , Receptors, Adrenergic, beta-2/metabolism , Rhodopsin/chemistry , Zinc/chemistry
10.
Bioorg Med Chem Lett ; 15(16): 3707-12, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-15993056

ABSTRACT

A computational protocol has been devised to relate 7TM receptor proteins (GPCRs) with respect to physicochemical features of the core ligand-binding site as defined from the crystal structure of bovine rhodopsin. The identification of such receptors that already are associated with ligand information (e.g., small molecule ligands with mutagenesis or SAR data) is used to support structure-guided drug design of novel ligands. A case targeting the newly identified prostaglandin D2 receptor CRTH2 serves as a primary example to illustrate the procedure.


Subject(s)
Computer Simulation , Drug Design , Hydrocarbons, Aromatic/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Rhodopsin/chemistry , Animals , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Binding Sites/physiology , Binding, Competitive/drug effects , Biphenyl Compounds , Cattle , Indomethacin/analogs & derivatives , Indomethacin/chemistry , Indomethacin/pharmacology , Ligands , Models, Biological , Molecular Structure , Receptors, G-Protein-Coupled/classification , Receptors, G-Protein-Coupled/metabolism , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/antagonists & inhibitors , Receptors, Prostaglandin/metabolism , Structure-Activity Relationship , Tetrazoles/chemistry , Tetrazoles/pharmacology
11.
Mol Pharmacol ; 68(2): 393-402, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15870392

ABSTRACT

The anti-inflammatory drugs indomethacin and ramatroban, the latter showing clinical efficacy in treating allergic asthma, have been shown to act as a classic agonist and antagonist, respectively, of the G protein-coupled chemoattractant receptor-homologous molecule expressed on Th2 cells (CRTH2 receptor). Here, we report the identification of two indole derivatives 1-(4-ethoxyphenyl)-5-methoxy-2-methylindole-3-carboxylic acid and N(alpha)-tosyltryptophan (hereafter referred to as 1 and 2, respectively), which are structurally related to indomethacin and ramatroban but which selectively interfere with a specific G protein-independent signaling pathway of CRTH2. In whole-cell saturation-binding assays, 1 and 2 both increase the number of [(3)H]prostaglandin D2 (PGD2)-recognizing CRTH2 sites and the affinity of PGD2 for CRTH2. Enzyme-linked immunosorbent assays show that they do not alter the total number of CRTH2 receptors on the cell surface. Analysis of their binding mode indicates that unlike indomethacin or ramatroban, 1 and 2 can occupy CRTH2 simultaneously with PGD2. On a functional level, however, 1 and 2 do not interfere with PGD2-mediated activation of heterotrimeric G proteins by CRTH2. In contrast, both compounds inhibit PGD2-mediated arrestin translocation via a G protein-independent mechanism. In human eosinophils endogenously expressing CRTH2, 1 selectively decreases the efficacy but not the potency of PGD2-induced shape change, unlike ramatroban, which displays competitive antagonistic behavior. These data show for the first time that "antagonists" can cause markedly dissimilar degrees of inhibition for different effector pathways and suggest that it may be possible to develop novel classes of specific signal-inhibiting drugs distinct from conventional antagonists.


Subject(s)
GTP-Binding Proteins/metabolism , Indoles/metabolism , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/metabolism , Signal Transduction/physiology , Animals , COS Cells , Chlorocebus aethiops , Dose-Response Relationship, Drug , Humans , Indoles/chemistry , Renilla
12.
J Virol ; 78(23): 12996-3006, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15542651

ABSTRACT

Here we report that the N-pyridinylmethyl cyclam analog AMD3451 has antiviral activity against a wide variety of R5, R5/X4, and X4 strains of human immunodeficiency virus type 1 (HIV-1) and HIV-2 (50% inhibitory concentration [IC(50)] ranging from 1.2 to 26.5 microM) in various T-cell lines, CCR5- or CXCR4-transfected cells, peripheral blood mononuclear cells (PBMCs), and monocytes/macrophages. AMD3451 also inhibited R5, R5/X4, and X4 HIV-1 primary clinical isolates in PBMCs (IC(50), 1.8 to 7.3 microM). A PCR-based viral entry assay revealed that AMD3451 blocks R5 and X4 HIV-1 infection at the virus entry stage. AMD3451 dose-dependently inhibited the intracellular Ca(2+) signaling induced by the CXCR4 ligand CXCL12 in T-lymphocytic cells and in CXCR4-transfected cells, as well as the Ca(2+) flux induced by the CCR5 ligands CCL5, CCL3, and CCL4 in CCR5-transfected cells. The compound did not interfere with chemokine-induced Ca(2+) signaling through CCR1, CCR2, CCR3, CCR4, CCR6, CCR9, or CXCR3 and did not induce intracellular Ca(2+) signaling by itself at concentrations up to 400 microM. In freshly isolated monocytes, AMD3451 inhibited the Ca(2+) flux induced by CXCL12 and CCL4 but not that induced by CCL2, CCL3, CCL5, and CCL7. The CXCL12- and CCL3-induced chemotaxis was also dose-dependently inhibited by AMD3451. Furthermore, AMD3451 inhibited CXCL12- and CCL3L1-induced endocytosis in CXCR4- and CCR5-transfected cells. AMD3451, in contrast to the specific CXCR4 antagonist AMD3100, did not inhibit but enhanced the binding of several anti-CXCR4 monoclonal antibodies (such as clone 12G5) at the cell surface, pointing to a different interaction with CXCR4. AMD3451 is the first low-molecular-weight anti-HIV agent with selective HIV coreceptor, CCR5 and CXCR4, interaction.


Subject(s)
Anti-HIV Agents/pharmacology , CCR5 Receptor Antagonists , HIV/drug effects , Heterocyclic Compounds/pharmacology , Pyridines/pharmacology , Receptors, CXCR4/antagonists & inhibitors , Virus Replication/drug effects , Antibodies, Monoclonal/immunology , Calcium/metabolism , Chemokine CXCL12 , Chemokines, CC/antagonists & inhibitors , Chemokines, CXC/antagonists & inhibitors , Chemotaxis/drug effects , HIV/physiology , Humans
13.
J Biol Chem ; 279(4): 3033-41, 2004 Jan 23.
Article in English | MEDLINE | ID: mdl-14585837

ABSTRACT

AMD3100 is a symmetric bicyclam, prototype non-peptide antagonist of the CXCR4 chemokine receptor. Mutational substitutions at 16 positions located in TM-III, -IV, -V, -VI, and -VII lining the main ligand-binding pocket of the CXCR4 receptor identified three acid residues: Asp(171) (AspIV:20), Asp(262) (AspVI:23), and Glu(288) (GluVII:06) as the main interaction points for AMD3100. Molecular modeling suggests that one cyclam ring of AMD3100 interacts with Asp(171) in TM-IV, whereas the other ring is sandwiched between the carboxylic acid groups of Asp(262) and Glu(288) from TM-VI and -VII, respectively. Metal ion binding in the cyclam rings of AMD3100 increased its dependence on Asp(262) and provided a tighter molecular map of the binding site, where borderline mutational hits became clear hits for the Zn(II)-loaded analog. The proposed binding site for AMD3100 was confirmed by a gradual build-up in the rather distinct CXCR3 receptor, for which the compound normally had no effect. Introduction of only a Glu at position VII:06 and the removal of a neutralizing Lys residue at position VII:02 resulted in a 1000-fold increase in affinity of AMD3100 to within 10-fold of its affinity in CXCR4. We conclude that AMD3100 binds through interactions with essentially only three acidic anchor-point residues, two of which are located at one end and the third at the opposite end of the main ligand-binding pocket of the CXCR4 receptor. We suggest that non-peptide antagonists with, for example, improved oral bioavailability can be designed to mimic this interaction and thereby efficiently and selectively block the CXCR4 receptor.


Subject(s)
Anti-HIV Agents/pharmacology , Heterocyclic Compounds/pharmacology , Receptors, CXCR4/antagonists & inhibitors , Anti-HIV Agents/chemistry , Benzylamines , Binding Sites , Cyclams , Heterocyclic Compounds/chemistry , Humans , Models, Molecular , Protein Binding , Protein Conformation , Receptors, CXCR4/chemistry , Signal Transduction
14.
Org Biomol Chem ; 1(23): 4248-53, 2003 Dec 07.
Article in English | MEDLINE | ID: mdl-14685327

ABSTRACT

With a view to probe the structure and function of G-protein coupled receptors the synthesis of functionalized 8-mercaptoquinoline derivatives and 2-(2-pyridyl)thiophenol was achieved. A fluorescence-based method for determining the affinity of these metal chelators toward zinc ions was developed.

15.
FEBS Lett ; 546(2-3): 300-6, 2003 Jul 10.
Article in English | MEDLINE | ID: mdl-12832058

ABSTRACT

The interaction of the CXCR4 antagonist AMD3100 with its target is greatly influenced by specific aspartate residues in the receptor protein, including Asp(171) and Asp(262). We have now found that aspartate-to-asparagine substitutions at these positions differentially affect the binding of four different anti-CXCR4 monoclonal antibodies as well as the infectivity of diverse human immunodeficiency virus type 1 (HIV-1) strains and clinical isolates. Mutation of Asp(262) strongly decreased the coreceptor efficiency of CXCR4 for wild-type but not for AMD3100-resistant HIV-1 NL4.3. Thus, resistance of HIV-1 NL4.3 to AMD3100 is associated with a decreased dependence of the viral gp120 on Asp(262) of CXCR4, pointing to a different mode of interaction of wild-type versus AMD3100-resistant virus with CXCR4.


Subject(s)
HIV-1/physiology , Heterocyclic Compounds/metabolism , Mutation , Receptors, CXCR4/metabolism , Amino Acid Sequence , Benzylamines , Binding Sites , Cyclams , Flow Cytometry , Humans , Membrane Fusion , Molecular Sequence Data , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/chemistry , Receptors, CXCR4/genetics , Receptors, CXCR4/immunology , Tumor Cells, Cultured
16.
Biochemistry ; 42(3): 710-7, 2003 Jan 28.
Article in English | MEDLINE | ID: mdl-12534283

ABSTRACT

The affinity of AMD3100, a symmetrical nonpeptide antagonist composed of two 1,4,8,11-tetraazacyclotetradecane (cyclam) rings connected through a 1,4-dimethylene(phenylene) linker to the CXCR4 chemokine receptor was increased 7, 36, and 50-fold, respectively, by incorporation of the following: Cu(2+), Zn(2+), or Ni(2+) into the cyclam rings of the compound. The rank order of the transition metal ions correlated with the calculated binding energy between free acetate and the metal ions coordinated in a cyclam ring. Construction of AMD3100 substituted with only a single Cu(2+) or Ni(2+) ion demonstrated that the increase in binding affinity of the metal ion substituted bicyclam is achieved through an enhanced interaction of just one of the ring systems. Mutational analysis of potential metal ion binding residues in the main ligand binding crevice of the CXCR4 receptor showed that although binding of the bicyclam is dependent on both Asp(171) and Asp(262), the enhancing effect of the metal ion was selectively eliminated by substitution of Asp(262) located at the extracellular end of TM-VI. It is concluded that the increased binding affinity of the metal ion substituted AMD3100 is obtained through enhanced interaction of one of the cyclam ring systems with the carboxylate group of Asp(262). It is suggested that this occurs through a strong concomitant interaction of one of the oxygen's directly with the metal ion and the other oxygen to one of the nitrogens of the cyclam ring through a hydrogen bond.


Subject(s)
Aspartic Acid/chemistry , Heterocyclic Compounds/chemistry , Metals, Heavy/chemistry , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/chemistry , Amino Acid Sequence , Animals , Aspartic Acid/genetics , Benzylamines , Binding, Competitive/genetics , COS Cells , Carboxylic Acids/chemistry , Cations, Divalent/chemistry , Chemokine CXCL12 , Chemokines, CXC/chemistry , Copper/chemistry , Cyclams , DNA Mutational Analysis , Humans , Ligands , Macromolecular Substances , Molecular Sequence Data , Mutagenesis, Site-Directed , Nickel/chemistry , Receptors, CXCR4/genetics , Transfection , Zinc/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...