Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Med Chem ; 55(11): 5311-25, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22563707

ABSTRACT

We present the systematic prospective evaluation of a protein-based and a ligand-based virtual screening platform against a set of three G-protein-coupled receptors (GPCRs): the ß-2 adrenoreceptor (ADRB2), the adenosine A(2A) receptor (AA2AR), and the sphingosine 1-phosphate receptor (S1PR1). Novel bioactive compounds were identified using a consensus scoring procedure combining ligand-based (frequent substructure ranking) and structure-based (Snooker) tools, and all 900 selected compounds were screened against all three receptors. A striking number of ligands showed affinity/activity for GPCRs other than the intended target, which could be partly attributed to the fuzziness and overlap of protein-based pharmacophore models. Surprisingly, the phosphodiesterase 5 (PDE5) inhibitor sildenafil was found to possess submicromolar affinity for AA2AR. Overall, this is one of the first published prospective chemogenomics studies that demonstrate the identification of novel cross-pharmacology between unrelated protein targets. The lessons learned from this study can be used to guide future virtual ligand design efforts.


Subject(s)
Databases, Factual , Drug Design , Models, Molecular , Quantitative Structure-Activity Relationship , Receptors, Adenosine A2/chemistry , Receptors, Adrenergic, beta-2/chemistry , Receptors, Lysosphingolipid/chemistry , Adenosine A2 Receptor Agonists/chemistry , Adenosine A2 Receptor Antagonists/chemistry , Adrenergic beta-2 Receptor Agonists/chemistry , Adrenergic beta-2 Receptor Antagonists/chemistry , Animals , CHO Cells , Cricetinae , Cricetulus , Drug Partial Agonism , HEK293 Cells , High-Throughput Screening Assays , Humans , Ligands , Molecular Structure , Phosphodiesterase 5 Inhibitors/chemistry , Piperazines/chemistry , Piperazines/metabolism , Purines/chemistry , Purines/metabolism , Radioligand Assay , Receptors, Adenosine A2/metabolism , Receptors, Adrenergic, beta-2/metabolism , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/metabolism , Sildenafil Citrate , Stochastic Processes , Sulfones/chemistry , Sulfones/metabolism
3.
Cell Host Microbe ; 8(6): 523-33, 2010 Dec 16.
Article in English | MEDLINE | ID: mdl-21147466

ABSTRACT

The WHIM syndrome, which features high susceptibility to human papillomavirus (HPV) infection, is a rare immunodeficiency associated with autosomal dominant heterozygous mutations of the CXCR4 chemokine receptor. CXCL12 and its receptors, CXCR4 and CXCR7, are linked to tumorigenesis, and we reported that abnormal expression of CXCL12 in epidermal keratinocytes correlates with HPV infection. However, the HPV-related pathologies observed in WHIM patients remain mechanistically unexplained. We show that keratinocytes immortalized by oncogenic HPV16 or HPV18 upregulate CXCL12 and its receptors in a manner dependent upon expression of the viral proteins E6 and E7. Autocrine signaling activated by CXCL12-engagement of its receptors controls motility and survival of the infected cells. Strikingly, expression of a WHIM syndrome-related gain-of-function CXCR4 mutant confers transforming capacity to HPV18-immortalized keratinocytes. These results establish a pivotal role for CXCL12 signaling in HPV-mediated transformation and provide a mechanistic basis for understanding HPV pathogenesis in WHIM syndrome.


Subject(s)
Chemokine CXCL12/physiology , Human papillomavirus 16/physiology , Human papillomavirus 18/physiology , Keratinocytes/virology , Papillomavirus Infections/virology , Receptors, CXCR/biosynthesis , Animals , Cell Movement , Cell Transformation, Viral , Chemokine CXCL12/biosynthesis , DNA-Binding Proteins/biosynthesis , Female , Humans , Immunologic Deficiency Syndromes/pathology , Immunologic Deficiency Syndromes/virology , Infant, Newborn , Male , Mice , Mice, Nude , Oncogene Proteins, Viral/biosynthesis , Papillomavirus E7 Proteins/biosynthesis , Papillomavirus Infections/pathology , Primary Immunodeficiency Diseases , Receptors, CXCR/physiology , Receptors, CXCR4/biosynthesis , Repressor Proteins/biosynthesis , Warts/pathology , Warts/virology
4.
Proc Natl Acad Sci U S A ; 107(47): 20565-70, 2010 Nov 23.
Article in English | MEDLINE | ID: mdl-21059953

ABSTRACT

The important family of G protein-coupled receptors has so far not been targeted very successfully with conventional monoclonal antibodies. Here we report the isolation and characterization of functional VHH-based immunoglobulin single variable domains (or nanobodies) against the chemokine receptor CXCR4. Two highly selective monovalent nanobodies, 238D2 and 238D4, were obtained using a time-efficient whole cell immunization, phage display, and counterselection method. The highly selective VHH-based immunoglobulin single variable domains competitively inhibited the CXCR4-mediated signaling and antagonized the chemoattractant effect of the CXCR4 ligand CXCL12. Epitope mapping showed that the two nanobodies bind to distinct but partially overlapping sites in the extracellular loops. Short peptide linkage of 238D2 with 238D4 resulted in significantly increased affinity for CXCR4 and picomolar activity in antichemotactic assays. Interestingly, the monovalent nanobodies behaved as neutral antagonists, whereas the biparatopic nanobodies acted as inverse agonists at the constitutively active CXCR4-N3.35A. The CXCR4 nanobodies displayed strong antiretroviral activity against T cell-tropic and dual-tropic HIV-1 strains. Moreover, the biparatopic nanobody effectively mobilized CD34-positive stem cells in cynomolgus monkeys. Thus, the nanobody platform may be highly effective at generating extremely potent and selective G protein-coupled receptor modulators.


Subject(s)
Antibodies/pharmacology , Chemotaxis/drug effects , HIV-1 , Receptors, CXCR4/immunology , Virus Replication/drug effects , Animals , Antibodies/isolation & purification , Antigens, CD34 , Benzylamines , Binding Sites/genetics , COS Cells , Chlorocebus aethiops , Cyclams , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , HEK293 Cells , Hematopoietic Stem Cell Mobilization , Heterocyclic Compounds , Humans , Reverse Transcriptase Polymerase Chain Reaction
5.
J Biol Chem ; 283(34): 23189-99, 2008 Aug 22.
Article in English | MEDLINE | ID: mdl-18556651

ABSTRACT

The chemokine CXCL12 and the receptor CXCR4 play pivotal roles in normal vascular and neuronal development, in inflammatory responses, and in infectious diseases and cancer. For instance, CXCL12 has been shown to mediate human immunodeficiency virus-induced neurotoxicity, proliferative retinopathy and chronic inflammation, whereas its receptor CXCR4 is involved in human immunodeficiency virus infection, cancer metastasis and in the rare disease known as the warts, hypogammaglobulinemia, immunodeficiency, and myelokathexis (WHIM) syndrome. As we screened chemical libraries to find inhibitors of the interaction between CXCL12 and the receptor CXCR4, we identified synthetic compounds from the family of chalcones that reduce binding of CXCL12 to CXCR4, inhibit calcium responses mediated by the receptor, and prevent CXCR4 internalization in response to CXCL12. We found that the chemical compounds display an original mechanism of action as they bind to the chemokine but not to CXCR4. The highest affinity molecule blocked chemotaxis of human peripheral blood lymphocytes ex vivo. It was also active in vivo in a mouse model of allergic eosinophilic airway inflammation in which we detected inhibition of the inflammatory infiltrate. The compound showed selectivity for CXCL12 and not for CCL5 and CXCL8 chemokines and blocked CXCL12 binding to its second receptor, CXCR7. By analogy to the effect of neutralizing antibodies, this molecule behaves as a small organic neutralizing compound that may prove to have valuable pharmacological and therapeutic potential.


Subject(s)
Chemokine CXCL12/metabolism , Gene Expression Regulation , Calcium/metabolism , Calorimetry , Cell Line , Cell Proliferation , Chalcones/metabolism , Chemokine CCL5/metabolism , Chemokine CXCL12/physiology , Humans , Inflammation , Ligands , Protein Binding , Receptors, CXCR/metabolism , Receptors, CXCR4/metabolism , Spectrometry, Fluorescence/methods
6.
Blood ; 112(1): 34-44, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18436740

ABSTRACT

WHIM (warts, hypogammaglobulinemia, infections, and myelokathexis) syndrome is an immune deficiency linked in many cases to heterozygous mutations causing truncations in the cytoplasmic tail of CXC chemokine receptor 4 (CXCR4). Leukocytes expressing truncated CXCR4 display enhanced responses to the receptor ligand CXCL12, including chemotaxis, which likely impair their trafficking and contribute to the immunohematologic clinical manifestations of the syndrome. CXCR4 desensitization and endocytosis are dependent on beta-arrestin (betaarr) recruitment to the cytoplasmic tail, so that the truncated CXCR4 are refractory to these processes and so have enhanced G protein-dependent signaling. Here, we show that the augmented responsiveness of WHIM leukocytes is also accounted for by enhanced betaarr2-dependent signaling downstream of the truncated CXCR4 receptor. Indeed, the WHIM-associated receptor CXCR4(1013) maintains association with betaarr2 and triggers augmented and prolonged betaarr2-dependent signaling, as revealed by ERK1/2 phosphorylation kinetics. Evidence is also provided that CXCR4(1013)-mediated chemotaxis critically requires betaarr2, and disrupting the SHSK motif in the third intracellular loop of CXCR4(1013) abrogates betaarr2-mediated signaling, but not coupling to G proteins, and normalizes chemotaxis. We also demonstrate that CXCR4(1013) spontaneously forms heterodimers with wild-type CXCR4. Accordingly, we propose a model where enhanced functional interactions between betaarr2 and receptor dimers account for the altered responsiveness of WHIM leukocytes to CXCL12.


Subject(s)
Arrestins/metabolism , Chemokine CXCL12/pharmacology , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/metabolism , Receptors, CXCR4/chemistry , Agammaglobulinemia/genetics , Agammaglobulinemia/immunology , Agammaglobulinemia/metabolism , Amino Acid Motifs , Cell Line , Chemotaxis, Leukocyte , Dimerization , Humans , Immunologic Deficiency Syndromes/genetics , Infections/genetics , Infections/immunology , Infections/metabolism , Mutation , Neutropenia/genetics , Neutropenia/immunology , Neutropenia/metabolism , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Signal Transduction , Warts/genetics , Warts/immunology , Warts/metabolism , beta-Arrestins
7.
Virology ; 371(1): 32-43, 2008 Feb 05.
Article in English | MEDLINE | ID: mdl-17961624

ABSTRACT

We have previously demonstrated that over-expression of spike protein (S) of severe acute respiratory syndrome coronavirus (SARS-CoV) or its C-terminal subunit (S2) is sufficient to induce apoptosis in vitro. To further investigate the possible roles of S2 in SARS-CoV-induced apoptosis and pathogenesis of SARS, we characterized the host expression profiles induced upon S2 over-expression in Vero E6 cells by oligonucleotide microarray analysis. Possible activation of mitochondrial apoptotic pathway in S2 expressing cells was suggested, as evidenced by the up-regulation of cytochrome c and down-regulation of the Bcl-2 family anti-apoptotic members. Inhibition of Bcl-2-related anti-apoptotic pathway was further supported by the diminution of S2-induced apoptosis in Vero E6 cells over-expressing Bcl-xL. In addition, modulation of CCN E2 and CDKN 1A implied the possible control of cell cycle arrest at G1/S phase. This study is expected to extend our understanding on the pathogenesis of SARS at a molecular level.


Subject(s)
Apoptosis , Cell Proliferation , Gene Expression Profiling , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Severe Acute Respiratory Syndrome/virology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Animals , Cell Line , Cell Survival , Chlorocebus aethiops , Gene Expression Regulation, Viral , Membrane Glycoproteins/genetics , Oligonucleotide Array Sequence Analysis , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Spike Glycoprotein, Coronavirus , Transcription, Genetic , Transduction, Genetic , Vero Cells , Viral Envelope Proteins/genetics
8.
J Biol Chem ; 282(8): 5111-5, 2007 Feb 23.
Article in English | MEDLINE | ID: mdl-17197449

ABSTRACT

Ligand binding to G protein-coupled receptors (GPCRs) is thought to induce changes in receptor conformation that translate into activation of downstream effectors. The link between receptor conformation and activity is still insufficiently understood, as current models of GPCR activation fail to take an increasing amount of experimental data into account. To elucidate structure-function relationships in GPCR activation, we used bioluminescence resonance energy transfer to directly assess the conformation of mutants of the chemokine receptor CXCR4. We analyzed substitutions in the arginine cage DRY motif and in the conserved asparagine N(3.35)119, which are pivotal molecular switches for receptor conformation and activation. G(alpha)(i) activation of the mutants was either similar to wild-type CXCR4 (D133N, Y135A, and N119D) or resulted in loss of activity (R134A and N119K). Mutant N119S was constitutively active but further activated by agonist. Bioluminescence resonance energy transfer analysis suggested no simple correlation between conformational changes in response to ligand binding and activation of G(alpha)(i) by the mutants. Different conformations of active receptors were detected (for wild-type CXCR4, D133N, and N119S), suggesting that different receptor conformations are able to trigger G(alpha)(i) activity. Several conformations were also found for inactive mutants. These data provide biophysical evidence for different receptor conformations being active with respect to a single readout. They support models of GPCR structure-activity relationships that take this conformational flexibility of active receptors into account.


Subject(s)
Amino Acid Substitution , GTP-Binding Protein alpha Subunits/metabolism , Models, Biological , Receptors, CXCR4/metabolism , Animals , Cell Line , Humans , Protein Conformation , Receptors, CXCR4/genetics , Signal Transduction/genetics
10.
FEBS Lett ; 579(30): 6699-704, 2005 Dec 19.
Article in English | MEDLINE | ID: mdl-16310778

ABSTRACT

The pro-apoptotic properties of severe acute respiratory syndrome coronavirus (SARS-CoV) structural proteins were studied in vitro. By monitoring apoptosis indicators including chromatin condensation, cellular DNA fragmentation and cell membrane asymmetry, we demonstrated that the adenovirus-mediated over-expression of SARS-CoV spike (S) protein and its C-terminal domain (S2) induce apoptosis in Vero E6 cells in a time- and dosage-dependent manner, whereas the expression of its N-terminal domain (S1) and other structural proteins, including envelope (E), membrane (M) and nucleocapsid (N) protein do not. These findings suggest a possible role of S and S2 protein in SARS-CoV induced apoptosis and the molecular pathogenesis of SARS.


Subject(s)
Adenoviridae/metabolism , Apoptosis , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Severe Acute Respiratory Syndrome/virology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Adenoviridae/genetics , Animals , Cell Line , Cell Survival , Chlorocebus aethiops , Genetic Vectors , Humans , Immunohistochemistry , Membrane Glycoproteins/genetics , Protein Structure, Tertiary , Spike Glycoprotein, Coronavirus , Time Factors , Transduction, Genetic , Vero Cells , Viral Envelope Proteins/genetics
11.
J Biol Chem ; 280(42): 35760-6, 2005 Oct 21.
Article in English | MEDLINE | ID: mdl-16107333

ABSTRACT

Combined phylogenetic and chromosomal location studies suggest that the orphan receptor RDC1 is related to CXC chemokine receptors. RDC1 provides a co-receptor function for a restricted number of human immunodeficiency virus (HIV) isolates, in particular for the CXCR4-using HIV-2 ROD strain. Here we show that CXCL12, the only known natural ligand for CXCR4, binds to and signals through RDC1. We demonstrate that RDC1 is expressed in T lymphocytes and that CXCL12-promoted chemotaxis is inhibited by an anti-RDC1 monoclonal antibody. Concomitant blockade of RDC1 and CXCR4 produced additive inhibitory effects in CXCL12-induced T cell migration. Furthermore, we provide evidence that interaction of CXCL12 with RDC1 is specific, saturable, and of high affinity (apparent KD approximately 0.4 nM). In CXCR4-negative cells expressing RDC1, CXCL12 promotes internalization of the receptor and chemotactic signals through RDC1. Collectively, our data indicate that RDC1, which we propose to rename as CXCR7, is a receptor for CXCL12.


Subject(s)
Chemokines, CXC/physiology , Receptors, G-Protein-Coupled/physiology , Signal Transduction , T-Lymphocytes/metabolism , Amino Acid Sequence , Antibodies, Monoclonal/chemistry , Biotinylation , Cell Line , Cell Movement , Chemokine CXCL12 , Chemokines, CXC/chemistry , Chemotaxis , Dose-Response Relationship, Drug , Down-Regulation , Electroporation , Flow Cytometry , Genetic Vectors , HIV/metabolism , Humans , Kinetics , Ligands , Microscopy, Fluorescence , Molecular Sequence Data , Receptors, CXCR , Receptors, G-Protein-Coupled/metabolism , Sequence Homology, Amino Acid , T-Lymphocytes/cytology , T-Lymphocytes/virology
SELECTION OF CITATIONS
SEARCH DETAIL