Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Med Chem ; 61(17): 7671-7686, 2018 09 13.
Article in English | MEDLINE | ID: mdl-30106292

ABSTRACT

We previously reported Chalcone-4 (1) that binds the chemokine CXCL12, not its cognate receptors CXCR4 or CXCR7, and neutralizes its biological activity. However, this neutraligand suffers from limitations such as poor chemical stability, solubility, and oral activity. Herein, we report on the discovery of pyrimidinone 57 (LIT-927), a novel neutraligand of CXCL12 which displays a higher solubility than 1 and is no longer a Michael acceptor. While both 1 and 57 reduce eosinophil recruitment in a murine model of allergic airway hypereosinophilia, 57 is the only one to display inhibitory activity following oral administration. Thereby, we here describe 57 as the first orally active CXCL12 neutraligand with anti-inflammatory properties. Combined with a high binding selectivity for CXCL12 over other chemokines, 57 represents a powerful pharmacological tool to investigate CXCL12 physiology in vivo and to explore the activity of chemokine neutralization in inflammatory and related diseases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Chemokine CXCL12/metabolism , Hypereosinophilic Syndrome/drug therapy , Pyrimidinones/chemistry , Pyrimidinones/pharmacology , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Chemokine CXCL12/chemistry , Disease Models, Animal , Drug Evaluation, Preclinical , Fluorescence Resonance Energy Transfer , Humans , Hypersensitivity/drug therapy , Hypersensitivity/etiology , Male , Mice, Inbred BALB C , Models, Molecular , Pyrimidinones/administration & dosage , Pyrimidinones/metabolism , Pyrimidinones/pharmacokinetics , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Structure-Activity Relationship
2.
Cell Rep ; 11(5): 737-47, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25921529

ABSTRACT

CXCL12-CXCR4 signaling controls multiple physiological processes and its dysregulation is associated with cancers and inflammatory diseases. To discover as-yet-unknown endogenous ligands of CXCR4, we screened a blood-derived peptide library for inhibitors of CXCR4-tropic HIV-1 strains. This approach identified a 16 amino acid fragment of serum albumin as an effective and highly specific CXCR4 antagonist. The endogenous peptide, termed EPI-X4, is evolutionarily conserved and generated from the highly abundant albumin precursor by pH-regulated proteases. EPI-X4 forms an unusual lasso-like structure and antagonizes CXCL12-induced tumor cell migration, mobilizes stem cells, and suppresses inflammatory responses in mice. Furthermore, the peptide is abundant in the urine of patients with inflammatory kidney diseases and may serve as a biomarker. Our results identify EPI-X4 as a key regulator of CXCR4 signaling and introduce proteolysis of an abundant precursor protein as an alternative concept for chemokine receptor regulation.


Subject(s)
Peptide Fragments/metabolism , Peptides/metabolism , Receptors, CXCR4/antagonists & inhibitors , Serum Albumin/metabolism , Amino Acid Sequence , Animals , Biomarkers/urine , Cell Line , Cell Movement/drug effects , HEK293 Cells , HIV-1/physiology , Half-Life , Humans , Jurkat Cells , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Peptide Library , Peptides/chemistry , Peptides/pharmacology , Protein Binding , Protein Structure, Tertiary , Receptors, CXCR4/metabolism , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Sequence Alignment , Serum Albumin/chemistry , Serum Albumin/pharmacology , Signal Transduction/drug effects , Virus Internalization/drug effects
3.
Mol Cancer ; 13: 58, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24629239

ABSTRACT

BACKGROUND: HIF-1α and CXCR4/CXCL12 have crucial roles in the metastatic process of colorectal cancer. Our aim was to study the significance of targeting HIF-1α and the CXCR4/CXCL12 axis in colorectal cancer to prevent the dissemination process in vitro. METHODS: We investigated CXCR4 and CXCR7 mRNA and protein expression in human colon carcinomas and the modulation of their expression by hypoxia and HIF-1α in colon cancer cell lines. The migration of tumor cells in a Boyden chamber was studied after CXCR4 inhibition with siRNA or the CXCR4/CXCL12 neutraligand, chalcone 4. RESULTS: Analysis of a cohort of colon polyps and chromosome-unstable carcinomas showed that the expression of CXCR4 and CXCR7 was similar to that of the normal mucosa in the polyps and early-stage carcinomas but significantly increased in late stage carcinomas. Our data demonstrate that hypoxia strongly induced the expression of CXCR4 transcript and protein at the cell membrane, both regulated by HIF-1α, whereas CXCR7 expression was independent of hypoxia. After transient hypoxia, CXCR4 levels remained stable at the cell membrane up to 48 hours. Furthermore, reducing CXCR4 expression impaired CXCL12-induced Akt phosphorylation, whereas Erk activation remained unchanged. In contrast, reducing CXCR7 expression did not affect Akt nor Erk activation. In the presence of CXCR4 or CXCR7 siRNAs, a significant reduction in cell migration occurred (37% and 17%, respectively). Although irinotecan inhibited cell migration by 20% (p <0.001), the irinotecan and chalcone 4 combination further increased inhibition to 40% (p <0.001). CONCLUSION: We demonstrated, for the first time, that hypoxia upregulated CXCR4 but not CXCR7 expression in tumor cells and that the CXCR4 receptor protein level remains high at the cell membrane when the tumor cells return to normoxia for up to 48 hours. In addition we showed the interest to inhibit the CXCR4 signaling by inhibiting both the HIF-1α and CXCR4/CXCL12 pathway. CXCR4 seems to be a relevant target because it is continuously expressed and functional both in normoxic and hypoxic conditions in tumor cells.


Subject(s)
Colonic Neoplasms/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Receptors, CXCR4/biosynthesis , Receptors, CXCR/biosynthesis , Cell Hypoxia/genetics , Cell Line, Tumor , Cell Movement/genetics , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptors, CXCR/genetics , Receptors, CXCR4/genetics , Signal Transduction
4.
J Cell Sci ; 127(Pt 4): 773-87, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24338366

ABSTRACT

The mTORC1 kinase promotes cell growth in response to growth factors by activation of receptor tyrosine kinase. It is regulated by the cellular energy level and the availability of nutrients. mTORC1 activity is also inhibited by cellular stresses through overexpression of REDD1 (regulated in development and DNA damage responses). We report the identification of REDD1 in a fluorescent live-imaging screen aimed at discovering new proteins implicated in G-protein-coupled receptor signaling, based on translocation criteria. Using a sensitive and quantitative plasma membrane localization assay based on bioluminescent resonance energy transfer, we further show that a panel of endogenously expressed GPCRs, through a Ca(2+)/calmodulin pathway, triggers plasma membrane translocation of REDD1 but not of its homolog REDD2. REDD1 and REDD2 share a conserved mTORC1-inhibitory motif characterized at the functional and structural level and differ most in their N-termini. We show that the N-terminus of REDD1 and its mTORC1-inhibitory motif participate in the GPCR-evoked dynamic interaction of REDD1 with the plasma membrane. We further identify REDD1 as a novel effector in GPCR signaling. We show that fast activation of mTORC1 by GPCRs correlates with fast and maximal translocation of REDD1 to the plasma membrane. Overexpression of functional REDD1 leads to a reduction of mTORC1 activation by GPCRs. By contrast, depletion of endogenous REDD1 protein unleashes mTORC1 activity. Thus, translocation to the plasma membrane appears to be an inactivation mechanism of REDD1 by GPCRs, which probably act by sequestering its functional mTORC1-inhibitory motif that is necessary for plasma membrane targeting.


Subject(s)
Cell Membrane/metabolism , Multiprotein Complexes/metabolism , Receptors, Neurokinin-2/metabolism , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Calcium Signaling , Calmodulin/metabolism , Enzyme Activation , HEK293 Cells , Humans , Mechanistic Target of Rapamycin Complex 1 , Molecular Sequence Data , Protein Interaction Domains and Motifs , Protein Sorting Signals , Protein Transport , Proteins/metabolism , Transcription Factors/chemistry
5.
J Biol Chem ; 288(17): 11865-76, 2013 Apr 26.
Article in English | MEDLINE | ID: mdl-23449983

ABSTRACT

The chemokine receptor CXCR4 and its chemokine CXCL12 are involved in normal tissue patterning but also in tumor cell growth and survival as well as in the recruitment of immune and inflammatory cells, as successfully demonstrated using agents that block either CXCL12 or CXCR4. In order to achieve selectivity in drug action on the CXCR4/CXCL12 pair, in particular in the airways, drugs should be delivered as selectively as possible in the treated tissue and should not diffuse in the systemic circulation, where it may reach undesired organs. To this end, we used a previously unexploited Knoevenagel reaction to create a short lived drug, or soft drug, based on the CXCL12-neutralizing small molecule, chalcone 4, which blocks binding of CXCL12 to CXCR4. We show that the compound, carbonitrile-chalcone 4, blocks the recruitment of eosinophils to the airways in ovalbumin-sensitized and challenged mice in vivo when administered directly to the airways by the intranasal route, but not when administered systemically by the intraperitoneal route. We show that the lack of effect at a distant site is due to the rapid degradation of the molecule to inactive fragments. This approach allows selective action of the CXCL12 neutraligands although the target protein is widely distributed in the organism.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Asthma/drug therapy , Chalcones/pharmacology , Chemokine CXCL12/antagonists & inhibitors , Animals , Anti-Asthmatic Agents/chemistry , Asthma/metabolism , Asthma/pathology , Chalcones/chemistry , Chemokine CXCL12/metabolism , Drug Evaluation, Preclinical , Eosinophils/metabolism , Eosinophils/pathology , HEK293 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Receptors, CXCR4/metabolism
6.
PLoS One ; 7(4): e34971, 2012.
Article in English | MEDLINE | ID: mdl-22511975

ABSTRACT

Litomosoides sigmodontis is a cause of filarial infection in rodents. Once infective larvae overcome the skin barrier, they enter the lymphatic system and then settle in the pleural cavity, causing soft tissue infection. The outcome of infection depends on the parasite's modulatory ability and also on the immune response of the infected host, which is influenced by its genetic background. The goal of this study was to determine whether host factors such as the chemokine axis CXCL12/CXCR4, which notably participates in the control of immune surveillance, can influence the outcome of the infection. We therefore set up comparative analyses of subcutaneous infection by L. sigmodontis in two inbred mouse strains with different outcomes: one susceptible strain (BALB/c) and one resistant strain (C57BL/6). We showed that rapid parasite clearance was associated with a L. sigmodontis-specific CXCL12-dependent cell response in C57BL/6 mice. CXCL12 was produced mainly by pleural mesothelial cells during infection. Conversely, the delayed parasite clearance in BALB/c mice was neither associated with an increase in CXCL12 levels nor with cell influx into the pleural cavity. Remarkably, interfering with the CXCL12/CXCR4 axis in both strains of mice delayed filarial development, as evidenced by the postponement of the fourth molting process. Furthermore, the in vitro growth of stage 4 filariae was favored by the addition of low amounts of CXCL12. The CXCL12/CXCR4 axis thus appears to have a dual effect on the L. sigmodontis life cycle: by acting as a host-cell restriction factor for infection, and as a growth factor for worms.


Subject(s)
Chemokine CXCL12/physiology , Filariasis/immunology , Animals , Filarioidea , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
7.
ACS Med Chem Lett ; 3(1): 10-4, 2012 Jan 12.
Article in English | MEDLINE | ID: mdl-24900366

ABSTRACT

Chalcone 4 (compound 1) is a small molecule that neutralizes the CXC chemokine CXCL12 and prevents it from acting on the CXCR4 and CXCR7 receptors. To overcome its poor solubility in aqueous buffers, we designed highly soluble analogues of compound 1, phosphate, l-seryl, and sulfate, all inactive by themselves on CXCL12 but when cleaved in vivo into 1, highly active locally at a low dose in a mouse airway hypereosinophilia model.

8.
Pharmacol Ther ; 126(1): 39-55, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20117133

ABSTRACT

Regulation of cellular responses to external stimuli such as hormones, neurotransmitters, or cytokines is achieved through the control of all steps of the complex cascade starting with synthesis, going through maturation steps, release, distribution, degradation and/or uptake of the signalling molecule interacting with the target protein. One possible way of regulation, referred to as scavenging or neutralization of the ligand, has been increasingly studied, especially for small protein ligands. It shows innovative potential in chemical biology approaches as well as in disease treatment. Neutralization of protein ligands, as for example cytokines or chemokines can lead to the validation of signalling pathways under physiological or pathophysiological conditions, and in certain cases, to the development of therapeutic molecules now used in autoimmune diseases, chronic inflammation and cancer treatment. This review explores the field of ligand neutralization and tries to determine to what extent small chemical molecules could substitute for neutralizing antibodies in therapeutic approaches.


Subject(s)
Antibodies, Neutralizing/therapeutic use , Chemokines/therapeutic use , Cytokines/therapeutic use , Inflammation/drug therapy , Chemokines/immunology , Chemokines/metabolism , Cytokines/immunology , Cytokines/metabolism , Humans , Models, Biological , Models, Molecular , Protein Binding , Signal Transduction/immunology , Signal Transduction/physiology
9.
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
10.
Traffic ; 9(3): 305-24, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18088318

ABSTRACT

Activated human neuropeptide Y Y(1) receptors rapidly desensitize and internalize through clathrin-coated pits and recycle from early and recycling endosomes, unlike Y(2) receptors that neither internalize nor desensitize. To identify motifs implicated in Y(1) receptor desensitization and trafficking, mutants with varying C-terminal truncations or a substituted Y(2) C-terminus were constructed. Point mutations of key putative residues were made in a C-terminal conserved motif [phi-H-(S/T)-(E/D)-V-(S/T)-X-T] that we have identified and in the second intracellular i2 loop. Receptors were analyzed by functional assays, spectrofluorimetric measurements on living cells, flow cytometry, confocal imaging and bioluminescence resonance energy transfer assays for beta-arrestin activation and adaptor protein (AP-2) complex recruitment. Inhibitory GTP-binding protein-dependent signaling of Y(1) receptors to adenylyl cyclase and desensitization was unaffected by C-terminal truncations or mutations, while C-terminal deletion mutants of 42 and 61 amino acids no longer internalized. Substitutions of Thr357, Asp358, Ser360 and Thr362 by Ala in the C-terminus abolished both internalization and beta-arrestin activation but not desensitization. A Pro145 substitution by His in an i2 consensus motif reported to mediate phosphorylation-independent recruitment of beta-arrestins affected neither desensitization, internalization or recycling kinetics of activated Y(1) receptors nor beta-arrestin activation. Interestingly, combining Pro145 substitution by His and C-terminal substitutions significantly attenuates Y(1) desensitization. In the Y(2) receptor, replacement of His155 with Pro at this position in the i2 loop motif promotes agonist-mediated desensitization, beta-arrestin activation, internalization and recycling. Overall, our results indicate that beta-arrestin-mediated desensitization and internalization of Y(1) and Y(2) receptors are differentially regulated by the C-terminal motif and the i2 loop consensus motif.


Subject(s)
Receptors, Neuropeptide Y/chemistry , Receptors, Neuropeptide Y/metabolism , Adenylyl Cyclases/metabolism , Amino Acid Motifs , Amino Acid Sequence , Amino Acid Substitution , Arrestins/metabolism , Biological Transport, Active , Cell Line , Cyclic AMP/metabolism , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Kinetics , Microscopy, Confocal , Molecular Sequence Data , Mutagenesis, Site-Directed , Receptors, Neuropeptide Y/agonists , Receptors, Neuropeptide Y/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Transfection , beta-Arrestins
11.
J Med Chem ; 50(6): 1294-303, 2007 Mar 22.
Article in English | MEDLINE | ID: mdl-17311371

ABSTRACT

A three-dimensional model of the chemokine receptor CCR5 has been built to fulfill structural peculiarities of its alpha-helix bundle and to distinguish known CCR5 antagonists from randomly chosen drug-like decoys. In silico screening of a library of 1.6 million commercially available compounds against the CCR5 model by sequential filters (drug-likeness, 2-D pharmacophore, 3-D docking, scaffold clustering) yielded a hit list of 59 compounds, out of which 10 exhibited a detectable binding affinity to the CCR5 receptor. Unexpectedly, most binders tested in a functional assay were shown to be agonists of the CCR5 receptor. A follow-up database query based on similarity to the most potent binders identified three new CCR5 agonists. Despite a moderate affinity of all nonpeptide ligands for the CCR5 receptor, one of the agonists was shown to promote efficient receptor internalization, which is a process therapeutically favorable for protection against HIV-1 infection.


Subject(s)
Models, Molecular , Quantitative Structure-Activity Relationship , Receptors, CCR5/agonists , Receptors, CCR5/chemistry , Amino Acid Sequence , CCR5 Receptor Antagonists , Molecular Sequence Data , Molecular Structure , Piperazines/chemistry , Piperidines/chemistry , Protein Structure, Tertiary , Pyridazines/chemistry , Pyrrolidines/chemistry , Quinazolines/chemistry , Sequence Homology, Amino Acid , Triazoles/chemistry
12.
Microsc Res Tech ; 69(12): 941-56, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17080432

ABSTRACT

Fluorescence resonance energy transfer (FRET) between an adequate pair of fluorophores is an indication of closer proximity than colocalization and is used by biologists to study fluorescently modified protein interactions inside cells. We present a method for visualization of FRET images acquired by confocal sensitized emission, involving excitation of the donor fluorophore and detection of the energy transfer as an emission from the acceptor fluorophore into the FRET channel. Authentic FRET signal measurements require the correction from the FRET channel of the undesired bleed-through signals (BT) resulting from both the leak-through of the donor emission and the direct acceptor emission. Our method reduces the interference of the user to a minimum by analyzing the entire image, pixel by pixel. It proposes imaging treatments and the display of control images to validate the BT calculation and the image corrections. It displays FRET images as a function of the colocalization of the two fluorescent partners. Finally, it proposes an alternative to normalization of the FRET intensities to compare FRET signal variations between samples. This method called "FRET and Colocalization Analyzer" has been implemented in a Plug-in of the freely available ImageJ software. It is particularly adapted when transient expression of the fluorescent proteins is used thereby giving very variable expression levels or when the colocalization of the two partners is varying in proportion, in amount, and in size, as a function of time. The method and program are validated using the analysis of the spatio-temporal interactions between a G-protein coupled receptor, the tachykinin NK2 receptor, and the beta-arrestin 2 as an example.


Subject(s)
Fluorescence Resonance Energy Transfer , Image Processing, Computer-Assisted , Microscopy, Confocal/methods , Arrestins/metabolism , Fluorescent Dyes , Humans , Receptors, G-Protein-Coupled/metabolism , Software , beta-Arrestin 2 , beta-Arrestins
SELECTION OF CITATIONS
SEARCH DETAIL
...