1
|
Endzhievskaya S, Chahal K, Resnick J, Khare E, Roy S, Handel TM, Kufareva I. Essential strategies for the detection of constitutive and ligand-dependent Gi-directed activity of 7TM receptors using bioluminescence resonance energy transfer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.04.626681. [PMID: 39713355 PMCID: PMC11661105 DOI: 10.1101/2024.12.04.626681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
The constitutive (ligand-independent) signaling of G protein-coupled receptors (GPCRs) is being increasingly appreciated as an integral aspect of their function; however, it can be technically hard to detect for poorly characterized, e.g. orphan, receptors of the cAMP-inhibitory Gi-coupled (GiPCR) family. In this study, we delineate the optimal strategies for the detection of such activity across several GiPCRs in two cell lines. As our study examples, we chose two canonical GiPCRs - the constitutively active Smoothened and the ligand-activated CXCR4, - and one atypical GPCRs, the chemokine receptor ACKR3. We verified the applicability of three Bioluminescence Resonance Energy Transfer (BRET)-based assays - one measuring changes in intracellular cAMP, another in Gβγ/GRK3ct association and third in Gαi-Gβγ dissociation, - for assessing both constitutive and ligand-modulated activity of these receptors. We also revealed the possible caveats and sources of false positives, and proposed optimization strategies. All three types of assays confirmed the ligand-dependent activity of CXCR4, the controversial G protein incompetence of ACKR3, the constitutive Gi-directed activity of SMO, and its modulation by PTCH1. We also demonstrated that PTCH1 promotes SMO localization to the cell surface, thus enhancing its responsiveness not only to agonists but also to antagonists, which is a novel mechanism of regulation of a Class F GiPCR Smoothened.
Collapse
Affiliation(s)
- Sofia Endzhievskaya
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Kirti Chahal
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
- LigronBio Inc., San Diego, CA, USA
| | - Julie Resnick
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Ekta Khare
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Suchismita Roy
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Tracy M. Handel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Irina Kufareva
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
2
|
Comerford I, McColl SR. Atypical chemokine receptors in the immune system. Nat Rev Immunol 2024; 24:753-769. [PMID: 38714818 DOI: 10.1038/s41577-024-01025-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2024] [Indexed: 05/10/2024]
Abstract
Leukocyte migration is a fundamental component of innate and adaptive immune responses as it governs the recruitment and localization of these motile cells, which is crucial for immune cell priming, effector functions, memory responses and immune regulation. This complex cellular trafficking system is controlled to a large extent via highly regulated production of secreted chemokines and the restricted expression of their membrane-tethered G-protein-coupled receptors. The activity of chemokines and their receptors is also regulated by a subfamily of molecules known as atypical chemokine receptors (ACKRs), which are chemokine receptor-like molecules that do not couple to the classical signalling pathways that promote cell migration in response to chemokine ligation. There has been a great deal of progress in understanding the biology of these receptors and their functions in the immune system in the past decade. Here, we describe the contribution of the various ACKRs to innate and adaptive immune responses, focussing specifically on recent progress. This includes recent findings that have defined the role for ACKRs in sculpting extracellular chemokine gradients, findings that broaden the spectrum of chemokine ligands recognized by these receptors, candidate new additions to ACKR family, and our increasing understanding of the role of these receptors in shaping the migration of innate and adaptive immune cells.
Collapse
Affiliation(s)
- Iain Comerford
- The Chemokine Biology Laboratory, School of Molecular & Biomedical Science, The University of Adelaide, Adelaide, South Australia, Australia.
| | - Shaun R McColl
- The Chemokine Biology Laboratory, School of Molecular & Biomedical Science, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
3
|
Otun O, Aljamous C, Del Nero E, Arimont-Segura M, Bosma R, Zarzycka B, Girbau T, Leyrat C, de Graaf C, Leurs R, Durroux T, Granier S, Cong X, Bechara C. Conformational dynamics underlying atypical chemokine receptor 3 activation. Proc Natl Acad Sci U S A 2024; 121:e2404000121. [PMID: 39008676 PMCID: PMC11287255 DOI: 10.1073/pnas.2404000121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 05/28/2024] [Indexed: 07/17/2024] Open
Abstract
Atypical Chemokine Receptor 3 (ACKR3) belongs to the G protein-coupled receptor family but it does not signal through G proteins. The structural properties that govern the functional selectivity and the conformational dynamics of ACKR3 activation are poorly understood. Here, we combined hydrogen/deuterium exchange mass spectrometry, site-directed mutagenesis, and molecular dynamics simulations to examine the binding mode and mechanism of action of ACKR3 ligands of different efficacies. Our results show that activation or inhibition of ACKR3 is governed by intracellular conformational changes of helix 6, intracellular loop 2, and helix 7, while the DRY motif becomes protected during both processes. Moreover, we identified the binding sites and the allosteric modulation of ACKR3 upon β-arrestin 1 binding. In summary, this study highlights the structure-function relationship of small ligands, the binding mode of β-arrestin 1, the activation dynamics, and the atypical dynamic features in ACKR3 that may contribute to its inability to activate G proteins.
Collapse
Affiliation(s)
- Omolade Otun
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier Cedex 534094, France
| | - Christelle Aljamous
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier Cedex 534094, France
| | - Elise Del Nero
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier Cedex 534094, France
| | - Marta Arimont-Segura
- Department of Medicinal Chemistry, Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam1081 HV, The Netherlands
| | - Reggie Bosma
- Department of Medicinal Chemistry, Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam1081 HV, The Netherlands
| | - Barbara Zarzycka
- Department of Medicinal Chemistry, Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam1081 HV, The Netherlands
| | - Tristan Girbau
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier Cedex 534094, France
| | - Cédric Leyrat
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier Cedex 534094, France
| | - Chris de Graaf
- Department of Medicinal Chemistry, Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam1081 HV, The Netherlands
| | - Rob Leurs
- Department of Medicinal Chemistry, Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam1081 HV, The Netherlands
| | - Thierry Durroux
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier Cedex 534094, France
| | - Sébastien Granier
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier Cedex 534094, France
| | - Xiaojing Cong
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier Cedex 534094, France
| | - Cherine Bechara
- Institut de Génomique Fonctionnelle, University of Montpellier, CNRS, INSERM, Montpellier Cedex 534094, France
- Institut Universitaire de France, Paris75005, France
| |
Collapse
|
4
|
Roy S, Sinha S, Silas AJ, Ghassemian M, Kufareva I, Ghosh P. Growth factor-dependent phosphorylation of Gα i shapes canonical signaling by G protein-coupled receptors. Sci Signal 2024; 17:eade8041. [PMID: 38833528 PMCID: PMC11328959 DOI: 10.1126/scisignal.ade8041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 05/17/2024] [Indexed: 06/06/2024]
Abstract
A long-standing question in the field of signal transduction is how distinct signaling pathways interact with each other to control cell behavior. Growth factor receptors and G protein-coupled receptors (GPCRs) are the two major signaling hubs in eukaryotes. Given that the mechanisms by which they signal independently have been extensively characterized, we investigated how they may cross-talk with each other. Using linear ion trap mass spectrometry and cell-based biophysical, biochemical, and phenotypic assays, we found at least three distinct ways in which epidermal growth factor affected canonical G protein signaling by the Gi-coupled GPCR CXCR4 through the phosphorylation of Gαi. Phosphomimicking mutations in two residues in the αE helix of Gαi (tyrosine-154/tyrosine-155) suppressed agonist-induced Gαi activation while promoting constitutive Gβγ signaling. Phosphomimicking mutations in the P loop (serine-44, serine-47, and threonine-48) suppressed Gi activation entirely, thus completely segregating growth factor and GPCR pathways. As expected, most of the phosphorylation events appeared to affect intrinsic properties of Gαi proteins, including conformational stability, nucleotide binding, and the ability to associate with and to release Gβγ. However, one phosphomimicking mutation, targeting the carboxyl-terminal residue tyrosine-320, promoted mislocalization of Gαi from the plasma membrane, a previously uncharacterized mechanism of suppressing GPCR signaling through G protein subcellular compartmentalization. Together, these findings elucidate not only how growth factor and chemokine signals cross-talk through the phosphorylation-dependent modulation of Gαi but also how such cross-talk may generate signal diversity.
Collapse
Affiliation(s)
- Suchismita Roy
- Department of Cellular and Molecular Medicine, University of California San Diego, CA 92093, USA
| | - Saptarshi Sinha
- Department of Cellular and Molecular Medicine, University of California San Diego, CA 92093, USA
| | - Ananta James Silas
- Department of Cellular and Molecular Medicine, University of California San Diego, CA 92093, USA
| | - Majid Ghassemian
- Department of Chemistry and Biochemistry, Biomolecular and Proteomics Mass Spectrometry Facility, University of California San Diego, San Diego, CA 92093, USA
| | - Irina Kufareva
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, CA 92093, USA
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California San Diego, CA 92093, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, CA 92093, USA
- Department of Medicine, University of California San Diego, CA 92093, USA
- Moore’s Comprehensive Cancer Center, University of California San Diego, CA 92093, USA
| |
Collapse
|
5
|
Zarca AM, Adlere I, Viciano CP, Arimont-Segura M, Meyrath M, Simon IA, Bebelman JP, Laan D, Custers HGJ, Janssen E, Versteegh KL, Buzink MCML, Nesheva DN, Bosma R, de Esch IJP, Vischer HF, Wijtmans M, Szpakowska M, Chevigné A, Hoffmann C, de Graaf C, Zarzycka BA, Windhorst AD, Smit MJ, Leurs R. Pharmacological Characterization and Radiolabeling of VUF15485, a High-Affinity Small-Molecule Agonist for the Atypical Chemokine Receptor ACKR3. Mol Pharmacol 2024; 105:301-312. [PMID: 38346795 DOI: 10.1124/molpharm.123.000835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/16/2024] [Indexed: 03/16/2024] Open
Abstract
Atypical chemokine receptor 3 (ACKR3), formerly referred to as CXCR7, is considered to be an interesting drug target. In this study, we report on the synthesis, pharmacological characterization and radiolabeling of VUF15485, a new ACKR3 small-molecule agonist, that will serve as an important new tool to study this β-arrestin-biased chemokine receptor. VUF15485 binds with nanomolar affinity (pIC50 = 8.3) to human ACKR3, as measured in [125I]CXCL12 competition binding experiments. Moreover, in a bioluminescence resonance energy transfer-based β-arrestin2 recruitment assay VUF15485 acts as a potent ACKR3 agonist (pEC50 = 7.6) and shows a similar extent of receptor activation compared with CXCL12 when using a newly developed, fluorescence resonance energy transfer-based ACKR3 conformational sensor. Moreover, the ACKR3 agonist VUF15485, tested against a (atypical) chemokine receptor panel (agonist and antagonist mode), proves to be selective for ACKR3. VUF15485 labeled with tritium at one of its methoxy groups ([3H]VUF15485), binds ACKR3 saturably and with high affinity (K d = 8.2 nM). Additionally, [3H]VUF15485 shows rapid binding kinetics and consequently a short residence time (<2 minutes) for binding to ACKR3. The selectivity of [3H]VUF15485 for ACKR3, was confirmed by binding studies, whereupon CXCR3, CXCR4, and ACKR3 small-molecule ligands were competed for binding against the radiolabeled agonist. Interestingly, the chemokine ligands CXCL11 and CXCL12 are not able to displace the binding of [3H]VUF15485 to ACKR3. The radiolabeled VUF15485 was subsequently used to evaluate its binding pocket. Site-directed mutagenesis and docking studies using a recently solved cryo-EM structure propose that VUF15485 binds in the major and the minor binding pocket of ACKR3. SIGNIFICANCE STATEMENT: The atypical chemokine receptor atypical chemokine receptor 3 (ACKR3) is considered an interesting drug target in relation to cancer and multiple sclerosis. The study reports on new chemical biology tools for ACKR3, i.e., a new agonist that can also be radiolabeled and a new ACKR3 conformational sensor, that both can be used to directly study the interaction of ACKR3 ligands with the G protein-coupled receptor.
Collapse
Affiliation(s)
- Aurelien M Zarca
- Department of Medicinal Chemistry (A.M.Z., M.A.-S., I.A.S., J.P.B., H.G.J.C., K.L.V., M.C.M.L.B., D.N.N., R.B., I.J.P.dE., H.F.V., M.W., C.dG., B.A.Z., M.J.S., R.L.) and Department of Chemistry & Pharmaceutical Sciences (E.J.), Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV Amsterdam, Netherlands; Griffin Discoveries BV, Amsterdam, Netherlands (I.A., I.J.P.dE., R.L.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum, Institut für Pharmakologie, Versbacher Strasse 9, 97078 Würzburg, Germany (C.P.V., C.H.); Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg (M.M., M.S., A.C.); and Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, Netherlands (D.L., A.D.W.)
| | - Ilze Adlere
- Department of Medicinal Chemistry (A.M.Z., M.A.-S., I.A.S., J.P.B., H.G.J.C., K.L.V., M.C.M.L.B., D.N.N., R.B., I.J.P.dE., H.F.V., M.W., C.dG., B.A.Z., M.J.S., R.L.) and Department of Chemistry & Pharmaceutical Sciences (E.J.), Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV Amsterdam, Netherlands; Griffin Discoveries BV, Amsterdam, Netherlands (I.A., I.J.P.dE., R.L.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum, Institut für Pharmakologie, Versbacher Strasse 9, 97078 Würzburg, Germany (C.P.V., C.H.); Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg (M.M., M.S., A.C.); and Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, Netherlands (D.L., A.D.W.)
| | - Cristina P Viciano
- Department of Medicinal Chemistry (A.M.Z., M.A.-S., I.A.S., J.P.B., H.G.J.C., K.L.V., M.C.M.L.B., D.N.N., R.B., I.J.P.dE., H.F.V., M.W., C.dG., B.A.Z., M.J.S., R.L.) and Department of Chemistry & Pharmaceutical Sciences (E.J.), Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV Amsterdam, Netherlands; Griffin Discoveries BV, Amsterdam, Netherlands (I.A., I.J.P.dE., R.L.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum, Institut für Pharmakologie, Versbacher Strasse 9, 97078 Würzburg, Germany (C.P.V., C.H.); Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg (M.M., M.S., A.C.); and Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, Netherlands (D.L., A.D.W.)
| | - Marta Arimont-Segura
- Department of Medicinal Chemistry (A.M.Z., M.A.-S., I.A.S., J.P.B., H.G.J.C., K.L.V., M.C.M.L.B., D.N.N., R.B., I.J.P.dE., H.F.V., M.W., C.dG., B.A.Z., M.J.S., R.L.) and Department of Chemistry & Pharmaceutical Sciences (E.J.), Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV Amsterdam, Netherlands; Griffin Discoveries BV, Amsterdam, Netherlands (I.A., I.J.P.dE., R.L.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum, Institut für Pharmakologie, Versbacher Strasse 9, 97078 Würzburg, Germany (C.P.V., C.H.); Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg (M.M., M.S., A.C.); and Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, Netherlands (D.L., A.D.W.)
| | - Max Meyrath
- Department of Medicinal Chemistry (A.M.Z., M.A.-S., I.A.S., J.P.B., H.G.J.C., K.L.V., M.C.M.L.B., D.N.N., R.B., I.J.P.dE., H.F.V., M.W., C.dG., B.A.Z., M.J.S., R.L.) and Department of Chemistry & Pharmaceutical Sciences (E.J.), Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV Amsterdam, Netherlands; Griffin Discoveries BV, Amsterdam, Netherlands (I.A., I.J.P.dE., R.L.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum, Institut für Pharmakologie, Versbacher Strasse 9, 97078 Würzburg, Germany (C.P.V., C.H.); Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg (M.M., M.S., A.C.); and Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, Netherlands (D.L., A.D.W.)
| | - Icaro A Simon
- Department of Medicinal Chemistry (A.M.Z., M.A.-S., I.A.S., J.P.B., H.G.J.C., K.L.V., M.C.M.L.B., D.N.N., R.B., I.J.P.dE., H.F.V., M.W., C.dG., B.A.Z., M.J.S., R.L.) and Department of Chemistry & Pharmaceutical Sciences (E.J.), Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV Amsterdam, Netherlands; Griffin Discoveries BV, Amsterdam, Netherlands (I.A., I.J.P.dE., R.L.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum, Institut für Pharmakologie, Versbacher Strasse 9, 97078 Würzburg, Germany (C.P.V., C.H.); Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg (M.M., M.S., A.C.); and Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, Netherlands (D.L., A.D.W.)
| | - Jan Paul Bebelman
- Department of Medicinal Chemistry (A.M.Z., M.A.-S., I.A.S., J.P.B., H.G.J.C., K.L.V., M.C.M.L.B., D.N.N., R.B., I.J.P.dE., H.F.V., M.W., C.dG., B.A.Z., M.J.S., R.L.) and Department of Chemistry & Pharmaceutical Sciences (E.J.), Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV Amsterdam, Netherlands; Griffin Discoveries BV, Amsterdam, Netherlands (I.A., I.J.P.dE., R.L.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum, Institut für Pharmakologie, Versbacher Strasse 9, 97078 Würzburg, Germany (C.P.V., C.H.); Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg (M.M., M.S., A.C.); and Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, Netherlands (D.L., A.D.W.)
| | - Dennis Laan
- Department of Medicinal Chemistry (A.M.Z., M.A.-S., I.A.S., J.P.B., H.G.J.C., K.L.V., M.C.M.L.B., D.N.N., R.B., I.J.P.dE., H.F.V., M.W., C.dG., B.A.Z., M.J.S., R.L.) and Department of Chemistry & Pharmaceutical Sciences (E.J.), Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV Amsterdam, Netherlands; Griffin Discoveries BV, Amsterdam, Netherlands (I.A., I.J.P.dE., R.L.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum, Institut für Pharmakologie, Versbacher Strasse 9, 97078 Würzburg, Germany (C.P.V., C.H.); Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg (M.M., M.S., A.C.); and Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, Netherlands (D.L., A.D.W.)
| | - Hans G J Custers
- Department of Medicinal Chemistry (A.M.Z., M.A.-S., I.A.S., J.P.B., H.G.J.C., K.L.V., M.C.M.L.B., D.N.N., R.B., I.J.P.dE., H.F.V., M.W., C.dG., B.A.Z., M.J.S., R.L.) and Department of Chemistry & Pharmaceutical Sciences (E.J.), Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV Amsterdam, Netherlands; Griffin Discoveries BV, Amsterdam, Netherlands (I.A., I.J.P.dE., R.L.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum, Institut für Pharmakologie, Versbacher Strasse 9, 97078 Würzburg, Germany (C.P.V., C.H.); Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg (M.M., M.S., A.C.); and Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, Netherlands (D.L., A.D.W.)
| | - Elwin Janssen
- Department of Medicinal Chemistry (A.M.Z., M.A.-S., I.A.S., J.P.B., H.G.J.C., K.L.V., M.C.M.L.B., D.N.N., R.B., I.J.P.dE., H.F.V., M.W., C.dG., B.A.Z., M.J.S., R.L.) and Department of Chemistry & Pharmaceutical Sciences (E.J.), Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV Amsterdam, Netherlands; Griffin Discoveries BV, Amsterdam, Netherlands (I.A., I.J.P.dE., R.L.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum, Institut für Pharmakologie, Versbacher Strasse 9, 97078 Würzburg, Germany (C.P.V., C.H.); Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg (M.M., M.S., A.C.); and Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, Netherlands (D.L., A.D.W.)
| | - Kobus L Versteegh
- Department of Medicinal Chemistry (A.M.Z., M.A.-S., I.A.S., J.P.B., H.G.J.C., K.L.V., M.C.M.L.B., D.N.N., R.B., I.J.P.dE., H.F.V., M.W., C.dG., B.A.Z., M.J.S., R.L.) and Department of Chemistry & Pharmaceutical Sciences (E.J.), Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV Amsterdam, Netherlands; Griffin Discoveries BV, Amsterdam, Netherlands (I.A., I.J.P.dE., R.L.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum, Institut für Pharmakologie, Versbacher Strasse 9, 97078 Würzburg, Germany (C.P.V., C.H.); Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg (M.M., M.S., A.C.); and Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, Netherlands (D.L., A.D.W.)
| | - Maurice C M L Buzink
- Department of Medicinal Chemistry (A.M.Z., M.A.-S., I.A.S., J.P.B., H.G.J.C., K.L.V., M.C.M.L.B., D.N.N., R.B., I.J.P.dE., H.F.V., M.W., C.dG., B.A.Z., M.J.S., R.L.) and Department of Chemistry & Pharmaceutical Sciences (E.J.), Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV Amsterdam, Netherlands; Griffin Discoveries BV, Amsterdam, Netherlands (I.A., I.J.P.dE., R.L.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum, Institut für Pharmakologie, Versbacher Strasse 9, 97078 Würzburg, Germany (C.P.V., C.H.); Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg (M.M., M.S., A.C.); and Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, Netherlands (D.L., A.D.W.)
| | - Desislava N Nesheva
- Department of Medicinal Chemistry (A.M.Z., M.A.-S., I.A.S., J.P.B., H.G.J.C., K.L.V., M.C.M.L.B., D.N.N., R.B., I.J.P.dE., H.F.V., M.W., C.dG., B.A.Z., M.J.S., R.L.) and Department of Chemistry & Pharmaceutical Sciences (E.J.), Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV Amsterdam, Netherlands; Griffin Discoveries BV, Amsterdam, Netherlands (I.A., I.J.P.dE., R.L.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum, Institut für Pharmakologie, Versbacher Strasse 9, 97078 Würzburg, Germany (C.P.V., C.H.); Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg (M.M., M.S., A.C.); and Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, Netherlands (D.L., A.D.W.)
| | - Reggie Bosma
- Department of Medicinal Chemistry (A.M.Z., M.A.-S., I.A.S., J.P.B., H.G.J.C., K.L.V., M.C.M.L.B., D.N.N., R.B., I.J.P.dE., H.F.V., M.W., C.dG., B.A.Z., M.J.S., R.L.) and Department of Chemistry & Pharmaceutical Sciences (E.J.), Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV Amsterdam, Netherlands; Griffin Discoveries BV, Amsterdam, Netherlands (I.A., I.J.P.dE., R.L.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum, Institut für Pharmakologie, Versbacher Strasse 9, 97078 Würzburg, Germany (C.P.V., C.H.); Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg (M.M., M.S., A.C.); and Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, Netherlands (D.L., A.D.W.)
| | - Iwan J P de Esch
- Department of Medicinal Chemistry (A.M.Z., M.A.-S., I.A.S., J.P.B., H.G.J.C., K.L.V., M.C.M.L.B., D.N.N., R.B., I.J.P.dE., H.F.V., M.W., C.dG., B.A.Z., M.J.S., R.L.) and Department of Chemistry & Pharmaceutical Sciences (E.J.), Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV Amsterdam, Netherlands; Griffin Discoveries BV, Amsterdam, Netherlands (I.A., I.J.P.dE., R.L.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum, Institut für Pharmakologie, Versbacher Strasse 9, 97078 Würzburg, Germany (C.P.V., C.H.); Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg (M.M., M.S., A.C.); and Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, Netherlands (D.L., A.D.W.)
| | - Henry F Vischer
- Department of Medicinal Chemistry (A.M.Z., M.A.-S., I.A.S., J.P.B., H.G.J.C., K.L.V., M.C.M.L.B., D.N.N., R.B., I.J.P.dE., H.F.V., M.W., C.dG., B.A.Z., M.J.S., R.L.) and Department of Chemistry & Pharmaceutical Sciences (E.J.), Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV Amsterdam, Netherlands; Griffin Discoveries BV, Amsterdam, Netherlands (I.A., I.J.P.dE., R.L.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum, Institut für Pharmakologie, Versbacher Strasse 9, 97078 Würzburg, Germany (C.P.V., C.H.); Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg (M.M., M.S., A.C.); and Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, Netherlands (D.L., A.D.W.)
| | - Maikel Wijtmans
- Department of Medicinal Chemistry (A.M.Z., M.A.-S., I.A.S., J.P.B., H.G.J.C., K.L.V., M.C.M.L.B., D.N.N., R.B., I.J.P.dE., H.F.V., M.W., C.dG., B.A.Z., M.J.S., R.L.) and Department of Chemistry & Pharmaceutical Sciences (E.J.), Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV Amsterdam, Netherlands; Griffin Discoveries BV, Amsterdam, Netherlands (I.A., I.J.P.dE., R.L.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum, Institut für Pharmakologie, Versbacher Strasse 9, 97078 Würzburg, Germany (C.P.V., C.H.); Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg (M.M., M.S., A.C.); and Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, Netherlands (D.L., A.D.W.)
| | - Martyna Szpakowska
- Department of Medicinal Chemistry (A.M.Z., M.A.-S., I.A.S., J.P.B., H.G.J.C., K.L.V., M.C.M.L.B., D.N.N., R.B., I.J.P.dE., H.F.V., M.W., C.dG., B.A.Z., M.J.S., R.L.) and Department of Chemistry & Pharmaceutical Sciences (E.J.), Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV Amsterdam, Netherlands; Griffin Discoveries BV, Amsterdam, Netherlands (I.A., I.J.P.dE., R.L.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum, Institut für Pharmakologie, Versbacher Strasse 9, 97078 Würzburg, Germany (C.P.V., C.H.); Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg (M.M., M.S., A.C.); and Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, Netherlands (D.L., A.D.W.)
| | - Andy Chevigné
- Department of Medicinal Chemistry (A.M.Z., M.A.-S., I.A.S., J.P.B., H.G.J.C., K.L.V., M.C.M.L.B., D.N.N., R.B., I.J.P.dE., H.F.V., M.W., C.dG., B.A.Z., M.J.S., R.L.) and Department of Chemistry & Pharmaceutical Sciences (E.J.), Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV Amsterdam, Netherlands; Griffin Discoveries BV, Amsterdam, Netherlands (I.A., I.J.P.dE., R.L.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum, Institut für Pharmakologie, Versbacher Strasse 9, 97078 Würzburg, Germany (C.P.V., C.H.); Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg (M.M., M.S., A.C.); and Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, Netherlands (D.L., A.D.W.)
| | - Carsten Hoffmann
- Department of Medicinal Chemistry (A.M.Z., M.A.-S., I.A.S., J.P.B., H.G.J.C., K.L.V., M.C.M.L.B., D.N.N., R.B., I.J.P.dE., H.F.V., M.W., C.dG., B.A.Z., M.J.S., R.L.) and Department of Chemistry & Pharmaceutical Sciences (E.J.), Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV Amsterdam, Netherlands; Griffin Discoveries BV, Amsterdam, Netherlands (I.A., I.J.P.dE., R.L.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum, Institut für Pharmakologie, Versbacher Strasse 9, 97078 Würzburg, Germany (C.P.V., C.H.); Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg (M.M., M.S., A.C.); and Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, Netherlands (D.L., A.D.W.)
| | - Chris de Graaf
- Department of Medicinal Chemistry (A.M.Z., M.A.-S., I.A.S., J.P.B., H.G.J.C., K.L.V., M.C.M.L.B., D.N.N., R.B., I.J.P.dE., H.F.V., M.W., C.dG., B.A.Z., M.J.S., R.L.) and Department of Chemistry & Pharmaceutical Sciences (E.J.), Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV Amsterdam, Netherlands; Griffin Discoveries BV, Amsterdam, Netherlands (I.A., I.J.P.dE., R.L.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum, Institut für Pharmakologie, Versbacher Strasse 9, 97078 Würzburg, Germany (C.P.V., C.H.); Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg (M.M., M.S., A.C.); and Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, Netherlands (D.L., A.D.W.)
| | - Barbara A Zarzycka
- Department of Medicinal Chemistry (A.M.Z., M.A.-S., I.A.S., J.P.B., H.G.J.C., K.L.V., M.C.M.L.B., D.N.N., R.B., I.J.P.dE., H.F.V., M.W., C.dG., B.A.Z., M.J.S., R.L.) and Department of Chemistry & Pharmaceutical Sciences (E.J.), Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV Amsterdam, Netherlands; Griffin Discoveries BV, Amsterdam, Netherlands (I.A., I.J.P.dE., R.L.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum, Institut für Pharmakologie, Versbacher Strasse 9, 97078 Würzburg, Germany (C.P.V., C.H.); Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg (M.M., M.S., A.C.); and Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, Netherlands (D.L., A.D.W.)
| | - Albert D Windhorst
- Department of Medicinal Chemistry (A.M.Z., M.A.-S., I.A.S., J.P.B., H.G.J.C., K.L.V., M.C.M.L.B., D.N.N., R.B., I.J.P.dE., H.F.V., M.W., C.dG., B.A.Z., M.J.S., R.L.) and Department of Chemistry & Pharmaceutical Sciences (E.J.), Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV Amsterdam, Netherlands; Griffin Discoveries BV, Amsterdam, Netherlands (I.A., I.J.P.dE., R.L.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum, Institut für Pharmakologie, Versbacher Strasse 9, 97078 Würzburg, Germany (C.P.V., C.H.); Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg (M.M., M.S., A.C.); and Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, Netherlands (D.L., A.D.W.)
| | - Martine J Smit
- Department of Medicinal Chemistry (A.M.Z., M.A.-S., I.A.S., J.P.B., H.G.J.C., K.L.V., M.C.M.L.B., D.N.N., R.B., I.J.P.dE., H.F.V., M.W., C.dG., B.A.Z., M.J.S., R.L.) and Department of Chemistry & Pharmaceutical Sciences (E.J.), Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV Amsterdam, Netherlands; Griffin Discoveries BV, Amsterdam, Netherlands (I.A., I.J.P.dE., R.L.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum, Institut für Pharmakologie, Versbacher Strasse 9, 97078 Würzburg, Germany (C.P.V., C.H.); Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg (M.M., M.S., A.C.); and Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, Netherlands (D.L., A.D.W.)
| | - Rob Leurs
- Department of Medicinal Chemistry (A.M.Z., M.A.-S., I.A.S., J.P.B., H.G.J.C., K.L.V., M.C.M.L.B., D.N.N., R.B., I.J.P.dE., H.F.V., M.W., C.dG., B.A.Z., M.J.S., R.L.) and Department of Chemistry & Pharmaceutical Sciences (E.J.), Amsterdam Institute for Molecular Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HV Amsterdam, Netherlands; Griffin Discoveries BV, Amsterdam, Netherlands (I.A., I.J.P.dE., R.L.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum, Institut für Pharmakologie, Versbacher Strasse 9, 97078 Würzburg, Germany (C.P.V., C.H.); Institute for Molecular Cell Biology, CMB - Center for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg (M.M., M.S., A.C.); and Department of Radiology and Nuclear Medicine, VU University Medical Center Amsterdam, Netherlands (D.L., A.D.W.)
| |
Collapse
|
6
|
Zerpa-Hernández DA, García-Chagollán M, Sánchez-Zuno GA, García-Arellano S, Hernández-Bello J, Hernández-Palma LA, Cerpa-Cruz S, Martinez-Bonilla G, Nicoletti F, Muñoz-Valle JF. Expression of Transcriptional Factors of T Helper Differentiation (T-bet, GATA-3, RORγt, and FOXP3), MIF Receptors (CD44, CD74, CXCR2, 4, 7), and Th1, Th2, and Th17 Cytokines in PBMC from Control Subjects and Rheumatoid Arthritis Patients. Curr Mol Med 2024; 24:1169-1182. [PMID: 37807647 DOI: 10.2174/0115665240260976230925095330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 08/09/2023] [Accepted: 08/11/2023] [Indexed: 10/10/2023]
Abstract
INTRODUCTION The macrophage migration inhibitory factor (MIF) plays a pivotal role in the development of rheumatoid arthritis (RA). Previous research indicates that MIF can trigger the expression of cytokine profiles associated with Th1, Th2, and Th17 responses in peripheral blood mononuclear cells (PBMC) from both RA patients and control subjects (CS). Despite these, few studies to date precisely elucidate the molecular mechanisms involved. The present study aimed to associate the expression of Th differentiation TF (T-bet, GATA-3, RORγt) with MIF receptors (CD44, CD74, CXCR2, 4, 7) and Th1, Th2, and Th17 cytokines in PBMC from CS and RA patients. METHOD PBMC from both groups was cultured for 24 h. The expression of the canonical and non-canonical MIF receptors and the TF was determined by flow cytometry. Additionally, multiplex bead analysis was employed to assess the levels of cytokines in the culture supernatants. The findings revealed that T CD4+ lymphocytes in the CS group exhibited a heightened expression of CD74 (p<0.05), whereas RA patients displayed an elevated expression of CXCR7 (p<0.001). Furthermore, T CD4+ lymphocytes from RA patients exhibited greater expression of GATA3, RORγt, and FOXP3, along with elevated levels of pro-inflammatory cytokines compared to the CS group (p<0.001). RESULT These results indicate that CD74 is more prominently expressed in PBMC from the CS group, whereas CXCR7 is more expressed in PBMC from RA patients. CONCLUSION We also noted an increased secretion of Th17 profile cytokines in RA, potentially influenced by the activation of FOXP3 via CD74 and RORγt through CXCR7 using the endocytic pathway.
Collapse
Affiliation(s)
| | - Mariel García-Chagollán
- Instituto de Investigación en Ciencias Biomédicas, Universidad de Guadalajara, Jalisco 44340, México
| | | | - Samuel García-Arellano
- Instituto de Investigación en Ciencias Biomédicas, Universidad de Guadalajara, Jalisco 44340, México
| | - Jorge Hernández-Bello
- Instituto de Investigación en Ciencias Biomédicas, Universidad de Guadalajara, Jalisco 44340, México
| | - Luis Alexis Hernández-Palma
- Instituto de Investigación en Ciencias Biomédicas, Universidad de Guadalajara, Jalisco 44340, México
- Instituto de Investigaciones en Comportamiento Alimentario y Nutrición, Universidad de Guadalajara, Jalisco 49000, Mexico
| | - Sergio Cerpa-Cruz
- Servicio de Reumatología, O.P.D. Hospital Civil de Guadalajara "Fray Antonio Alcalde", Jalisco 44280, Mexico
| | - Gloria Martinez-Bonilla
- Servicio de Reumatología, O.P.D. Hospital Civil de Guadalajara "Fray Antonio Alcalde", Jalisco 44280, Mexico
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | | |
Collapse
|
7
|
Gonçalves TL, de Araújo LP, Pereira Ferrer V. Tamoxifen as a modulator of CXCL12-CXCR4-CXCR7 chemokine axis: A breast cancer and glioblastoma view. Cytokine 2023; 170:156344. [PMID: 37639844 DOI: 10.1016/j.cyto.2023.156344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/18/2023] [Accepted: 08/20/2023] [Indexed: 08/31/2023]
Abstract
The chemokine stromal cell-derived-factor 1 (SDF)-1/CXCL12 acts by binding to its receptors, the CXC-4 chemokine receptor (CXCR4) and the CXC-7 chemokine receptor (CXCR7). The binding of CXCL12 to its receptors results in downstream signaling that leads to cell survival, proliferation and migration of tumor cells. CXCL12 and CXCR4 are highly expressed in breast cancer (BC) and glioblastoma (GBM) compared to normal cells. High expression of this chemokine axis correlates with increased therapy resistance and grade, tumor spread and poorer prognosis in these tumors. Tamoxifen (TMX) is a selective estrogen receptor modulator (SERM) that inhibits the expression of estrogen-regulated genes, including growth and angiogenic factors secreted by tumor cells. Additionally, TMX targets several proteins, such as protein kinase C (PKC), phospholipase C (PLC), P-glycoprotein (PgP), phosphatidylinositol-3-kinase (PI3K) and ion channels. This drug showed promising antitumor activity against both BC and GBM cells. In this review, we discuss the role of the CXCL12-CXCR4-CXCR7 chemokine axis in BC and GBM tumor biology and propose TMX as a potential modulator of this axis in these tumors. TMX modulates the CXCL12-CXCR4-CXCR7 axis in BC, however, there are no studies on this in GBM. We propose that studying this axis in GBM cells/patients treated with TMX might be beneficial for these patients. TMX inhibits important signaling pathways in these tumors and the activation of this chemokine axis is associated with increased therapy resistance.
Collapse
Affiliation(s)
- Thaynan Lopes Gonçalves
- Laboratory of Cell and Molecular Biology of Tumors, Department of Cell and Molecular Biology, Biology Institute, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil; Postgraduate Program in Pathological Anatomy, Faculty of Medicine, Rio de Janeiro Federal University, Rio de Janeiro, Brazil
| | - Luanna Prudencio de Araújo
- Laboratory of Cell and Molecular Biology of Tumors, Department of Cell and Molecular Biology, Biology Institute, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil
| | - Valéria Pereira Ferrer
- Laboratory of Cell and Molecular Biology of Tumors, Department of Cell and Molecular Biology, Biology Institute, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil; Postgraduate Program in Pathological Anatomy, Faculty of Medicine, Rio de Janeiro Federal University, Rio de Janeiro, Brazil.
| |
Collapse
|
8
|
Schafer CT, Chen Q, Tesmer JJG, Handel TM. Atypical Chemokine Receptor 3 "Senses" CXC Chemokine Receptor 4 Activation Through GPCR Kinase Phosphorylation. Mol Pharmacol 2023; 104:174-186. [PMID: 37474305 PMCID: PMC11033958 DOI: 10.1124/molpharm.123.000710] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/22/2023] [Accepted: 06/30/2023] [Indexed: 07/22/2023] Open
Abstract
Atypical chemokine receptor 3 (ACKR3) is an arrestin-biased receptor that regulates extracellular chemokine levels through scavenging. The scavenging process restricts the availability of the chemokine agonist CXCL12 for the G protein-coupled receptor (GPCR) CXCR4 and requires phosphorylation of the ACKR3 C-terminus by GPCR kinases (GRKs). ACKR3 is phosphorylated by GRK2 and GRK5, but the mechanisms by which these kinases regulate the receptor are unresolved. Here we determined that GRK5 phosphorylation of ACKR3 results in more efficient chemokine scavenging and β-arrestin recruitment than phosphorylation by GRK2 in HEK293 cells. However, co-activation of CXCR4-enhanced ACKR3 phosphorylation by GRK2 through the liberation of Gβγ, an accessory protein required for efficient GRK2 activity. The results suggest that ACKR3 "senses" CXCR4 activation through a GRK2-dependent crosstalk mechanism, which enables CXCR4 to influence the efficiency of CXCL12 scavenging and β-arrestin recruitment to ACKR3. Surprisingly, we also found that despite the requirement for phosphorylation and the fact that most ligands promote β-arrestin recruitment, β-arrestins are dispensable for ACKR3 internalization and scavenging, suggesting a yet-to-be-determined function for these adapter proteins. Since ACKR3 is also a receptor for CXCL11 and opioid peptides, these data suggest that such crosstalk may also be operative in cells with CXCR3 and opioid receptor co-expression. Additionally, kinase-mediated receptor cross-regulation may be relevant to other atypical and G protein-coupled receptors that share common ligands. SIGNIFICANCE STATEMENT: The atypical receptor ACKR3 indirectly regulates CXCR4-mediated cell migration by scavenging their shared agonist CXCL12. Here, we show that scavenging and β-arrestin recruitment by ACKR3 are primarily dependent on phosphorylation by GRK5. However, we also show that CXCR4 co-activation enhances the contribution of GRK2 by liberating Gβγ. This phosphorylation crosstalk may represent a common feedback mechanism between atypical and G protein-coupled receptors with shared ligands for regulating the efficiency of scavenging or other atypical receptor functions.
Collapse
Affiliation(s)
- Christopher T Schafer
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California (C.T.S., T.M.H.) and Departments of Biological Sciences and of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (Q.C., J.J.G.T.)
| | - Qiuyan Chen
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California (C.T.S., T.M.H.) and Departments of Biological Sciences and of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (Q.C., J.J.G.T.)
| | - John J G Tesmer
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California (C.T.S., T.M.H.) and Departments of Biological Sciences and of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (Q.C., J.J.G.T.)
| | - Tracy M Handel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California (C.T.S., T.M.H.) and Departments of Biological Sciences and of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (Q.C., J.J.G.T.)
| |
Collapse
|
9
|
Chen Q, Schafer CT, Mukherjee S, Gustavsson M, Agrawal P, Yao XQ, Kossiakoff AA, Handel TM, Tesmer JJG. ACKR3-arrestin2/3 complexes reveal molecular consequences of GRK-dependent barcoding. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.18.549504. [PMID: 37502840 PMCID: PMC10370059 DOI: 10.1101/2023.07.18.549504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Atypical chemokine receptor 3 (ACKR3, also known as CXCR7) is a scavenger receptor that regulates extracellular levels of the chemokine CXCL12 to maintain responsiveness of its partner, the G protein-coupled receptor (GPCR), CXCR4. ACKR3 is notable because it does not couple to G proteins and instead is completely biased towards arrestins. Our previous studies revealed that GRK2 and GRK5 install distinct distributions of phosphates (or "barcodes") on the ACKR3 carboxy terminal tail, but how these unique barcodes drive different cellular outcomes is not understood. It is also not known if arrestin2 (Arr2) and 3 (Arr3) bind to these barcodes in distinct ways. Here we report cryo-electron microscopy structures of Arr2 and Arr3 in complex with ACKR3 phosphorylated by either GRK2 or GRK5. Unexpectedly, the finger loops of Arr2 and 3 directly insert into the detergent/membrane instead of the transmembrane core of ACKR3, in contrast to previously reported "core" GPCR-arrestin complexes. The distance between the phosphorylation barcode and the receptor transmembrane core regulates the interaction mode of arrestin, alternating between a tighter complex for GRK5 sites and heterogenous primarily "tail only" complexes for GRK2 sites. Arr2 and 3 bind at different angles relative to the core of ACKR3, likely due to differences in membrane/micelle anchoring at their C-edge loops. Our structural investigations were facilitated by Fab7, a novel Fab that binds both Arr2 and 3 in their activated states irrespective of receptor or phosphorylation status, rendering it a potentially useful tool to aid structure determination of any native GPCR-arrestin complex. The structures provide unprecedented insight into how different phosphorylation barcodes and arrestin isoforms can globally affect the configuration of receptor-arrestin complexes. These differences may promote unique downstream intracellular interactions and cellular responses. Our structures also suggest that the 100% bias of ACKR3 for arrestins is driven by the ability of arrestins, but not G proteins, to bind GRK-phosphorylated ACKR3 even when excluded from the receptor cytoplasmic binding pocket.
Collapse
Affiliation(s)
- Qiuyan Chen
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biological Sciences, Purdue University, West Lafayette IN 47907-2054, USA
| | - Christopher T Schafer
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093 USA
- Department of Medicinal Chemistry, Amsterdam Institute for Molecular and Life Sciences, Faculty of Science, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, Netherlands
| | - Somnath Mukherjee
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL60637, USA
| | - Martin Gustavsson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093 USA
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Parth Agrawal
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL60637, USA
| | - Xin-Qiu Yao
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA
| | - Anthony A Kossiakoff
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL60637, USA
| | - Tracy M Handel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093 USA
| | - John J G Tesmer
- Department of Biological Sciences, Purdue University, West Lafayette IN 47907-2054, USA
| |
Collapse
|
10
|
Schafer CT, Chen Q, Tesmer JJG, Handel TM. Atypical Chemokine Receptor 3 'Senses' CXC Chemokine Receptor 4 Activation Through GPCR Kinase Phosphorylation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.25.530029. [PMID: 36865154 PMCID: PMC9980177 DOI: 10.1101/2023.02.25.530029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2023]
Abstract
Atypical chemokine receptor 3 (ACKR3) is an arrestin-biased receptor that regulates extracellular chemokine levels through scavenging. The scavenging action mediates the availability of the chemokine CXCL12 for the G protein-coupled receptor (GPCR) CXCR4 and requires phosphorylation of the ACKR3 C-terminus by GPCR kinases (GRKs). ACKR3 is phosphorylated by GRK2 and GRK5, but the mechanisms by which these kinases regulate the receptor are unresolved. Here we mapped the phosphorylation patterns and determined that GRK5 phosphorylation of ACKR3 dominates β-arrestin recruitment and chemokine scavenging over GRK2. Co-activation of CXCR4 significantly enhanced phosphorylation by GRK2 through the liberation of Gβγ. These results suggest that ACKR3 'senses' CXCR4 activation through a GRK2-dependent crosstalk mechanism. Surprisingly, we also found that despite the requirement for phosphorylation, and the fact that most ligands promote β-arrestin recruitment, β-arrestins are dispensable for ACKR3 internalization and scavenging, suggesting a yet to be determined function for these adapter proteins.
Collapse
Affiliation(s)
- Christopher T. Schafer
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, USA
| | - Qiuyan Chen
- Departments of Biological Sciences and of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
- Present address: Dept. of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - John J. G. Tesmer
- Departments of Biological Sciences and of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - Tracy M. Handel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, USA
| |
Collapse
|
11
|
Szpakowska M, D’Uonnolo G, Luís R, Alonso Bartolomé A, Thelen M, Legler DF, Chevigné A. New pairings and deorphanization among the atypical chemokine receptor family - physiological and clinical relevance. Front Immunol 2023; 14:1133394. [PMID: 37153591 PMCID: PMC10157204 DOI: 10.3389/fimmu.2023.1133394] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 03/30/2023] [Indexed: 05/09/2023] Open
Abstract
Atypical chemokine receptors (ACKRs) form a small subfamily of receptors (ACKR1-4) unable to trigger G protein-dependent signaling in response to their ligands. They do, however, play a crucial regulatory role in chemokine biology by capturing, scavenging or transporting chemokines, thereby regulating their availability and signaling through classical chemokine receptors. ACKRs add thus another layer of complexity to the intricate chemokine-receptor interaction network. Recently, targeted approaches and screening programs aiming at reassessing chemokine activity towards ACKRs identified several new pairings such as the dimeric CXCL12 with ACKR1, CXCL2, CXCL10 and CCL26 with ACKR2, the viral broad-spectrum chemokine vCCL2/vMIP-II, a range of opioid peptides and PAMP-12 with ACKR3 as well as CCL20 and CCL22 with ACKR4. Moreover, GPR182 (ACKR5) has been lately proposed as a new promiscuous atypical chemokine receptor with scavenging activity notably towards CXCL9, CXCL10, CXCL12 and CXCL13. Altogether, these findings reveal new degrees of complexity of the chemokine network and expand the panel of ACKR ligands and regulatory functions. In this minireview, we present and discuss these new pairings, their physiological and clinical relevance as well as the opportunities they open for targeting ACKRs in innovative therapeutic strategies.
Collapse
Affiliation(s)
- Martyna Szpakowska
- Immuno-Pharmacology and Interactomics,Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Giulia D’Uonnolo
- Immuno-Pharmacology and Interactomics,Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Rafael Luís
- Immuno-Pharmacology and Interactomics,Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Tumor Immunotherapy and Microenvironment, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Ana Alonso Bartolomé
- Immuno-Pharmacology and Interactomics,Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Marcus Thelen
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Daniel F. Legler
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, Kreuzlingen, Switzerland
| | - Andy Chevigné
- Immuno-Pharmacology and Interactomics,Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- *Correspondence: Andy Chevigné,
| |
Collapse
|
12
|
Yen YC, Schafer CT, Gustavsson M, Eberle SA, Dominik PK, Deneka D, Zhang P, Schall TJ, Kossiakoff AA, Tesmer JJG, Handel TM. Structures of atypical chemokine receptor 3 reveal the basis for its promiscuity and signaling bias. SCIENCE ADVANCES 2022; 8:eabn8063. [PMID: 35857509 PMCID: PMC9278869 DOI: 10.1126/sciadv.abn8063] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 05/26/2022] [Indexed: 05/12/2023]
Abstract
Both CXC chemokine receptor 4 (CXCR4) and atypical chemokine receptor 3 (ACKR3) are activated by the chemokine CXCL12 yet evoke distinct cellular responses. CXCR4 is a canonical G protein-coupled receptor (GPCR), whereas ACKR3 is intrinsically biased for arrestin. The molecular basis for this difference is not understood. Here, we describe cryo-EM structures of ACKR3 in complex with CXCL12, a more potent CXCL12 variant, and a small-molecule agonist. The bound chemokines adopt an unexpected pose relative to those established for CXCR4 and observed in other receptor-chemokine complexes. Along with functional studies, these structures provide insight into the ligand-binding promiscuity of ACKR3, why it fails to couple to G proteins, and its bias toward β-arrestin. The results lay the groundwork for understanding the physiological interplay of ACKR3 with other GPCRs.
Collapse
Affiliation(s)
- Yu-Chen Yen
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Christopher T. Schafer
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Martin Gustavsson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Stefanie A. Eberle
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Pawel K. Dominik
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Dawid Deneka
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
- Department of Biophysics, Jagiellonian University, Krakow, Poland
| | - Penglie Zhang
- ChemoCentryx Inc., 835 Industrial Rd., Suite 600, San Carlos, CA 94070, USA
| | - Thomas J. Schall
- ChemoCentryx Inc., 835 Industrial Rd., Suite 600, San Carlos, CA 94070, USA
| | - Anthony A. Kossiakoff
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - John J. G. Tesmer
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
- Department of Molecular Pharmacology and Medicinal Chemistry, Purdue University, West Lafayette, IN, USA
| | - Tracy M. Handel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
13
|
D'Uonnolo G, Reynders N, Meyrath M, Abboud D, Uchański T, Laeremans T, Volkman BF, Janji B, Hanson J, Szpakowska M, Chevigné A. The Extended N-Terminal Domain Confers Atypical Chemokine Receptor Properties to CXCR3-B. Front Immunol 2022; 13:868579. [PMID: 35720349 PMCID: PMC9198273 DOI: 10.3389/fimmu.2022.868579] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 04/15/2022] [Indexed: 11/24/2022] Open
Abstract
The chemokine receptor CXCR3 plays a critical role in immune cell recruitment and activation. CXCR3 exists as two main isoforms, CXCR3-A and CXCR3-B, resulting from alternative splicing. Although the two isoforms differ only by the presence of an N-terminal extension in CXCR3-B, they have been attributed divergent functional effects on cell migration and proliferation. CXCR3-B is the more enigmatic isoform and the mechanisms underlying its function and signaling remain elusive. We therefore undertook an in-depth cellular and molecular comparative study of CXCR3-A and CXCR3-B, investigating their activation at different levels of the signaling cascades, including G protein coupling, β-arrestin recruitment and modulation of secondary messengers as well as their downstream gene response elements. We also compared the subcellular localization of the two isoforms and their trafficking under resting and stimulated conditions along with their ability to internalize CXCR3-related chemokines. Here, we show that the N-terminal extension of CXCR3-B drastically affects receptor features, modifying its cellular localization and preventing G protein coupling, while preserving β-arrestin recruitment and chemokine uptake capacities. Moreover, we demonstrate that gradual truncation of the N terminus leads to progressive recovery of surface expression and G protein coupling. Our study clarifies the molecular basis underlying the divergent effects of CXCR3 isoforms, and emphasizes the β-arrestin-bias and the atypical nature of CXCR3-B.
Collapse
Affiliation(s)
- Giulia D'Uonnolo
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.,Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Nathan Reynders
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.,Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Max Meyrath
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Dayana Abboud
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases, University of Liège, Liège, Belgium
| | - Tomasz Uchański
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | | | - Brian F Volkman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Bassam Janji
- Tumor Immunotherapy and Microenvironment, Department of Oncology, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Julien Hanson
- Laboratory of Molecular Pharmacology, GIGA-Molecular Biology of Diseases, University of Liège, Liège, Belgium.,Laboratory of Medicinal Chemistry, Centre for Interdisciplinary Research on Medicines (CIRM), University of Liège, Liège, Belgium
| | - Martyna Szpakowska
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.,Tumor Immunotherapy and Microenvironment, Department of Oncology, Luxembourg Institute of Health, Luxembourg City, Luxembourg
| | - Andy Chevigné
- Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
14
|
Eberle SA, Gustavsson M. A Scintillation Proximity Assay for Real-Time Kinetic Analysis of Chemokine-Chemokine Receptor Interactions. Cells 2022; 11:1317. [PMID: 35455996 PMCID: PMC9024993 DOI: 10.3390/cells11081317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/05/2022] [Accepted: 04/11/2022] [Indexed: 11/23/2022] Open
Abstract
Chemokine receptors are extensively involved in a broad range of physiological and pathological processes, making them attractive drug targets. However, despite considerable efforts, there are very few approved drugs targeting this class of seven transmembrane domain receptors to date. In recent years, the importance of including binding kinetics in drug discovery campaigns was emphasized. Therefore, kinetic insight into chemokine-chemokine receptor interactions could help to address this issue. Moreover, it could additionally deepen our understanding of the selectivity and promiscuity of the chemokine-chemokine receptor network. Here, we describe the application, optimization and validation of a homogenous Scintillation Proximity Assay (SPA) for real-time kinetic profiling of chemokine-chemokine receptor interactions on the example of ACKR3 and CXCL12. The principle of the SPA is the detection of radioligand binding to receptors reconstituted into nanodiscs by scintillation light. No receptor modifications are required. The nanodiscs provide a native-like environment for receptors and allow for full control over bilayer composition and size. The continuous assay format enables the monitoring of binding reactions in real-time, and directly accounts for non-specific binding and potential artefacts. Minor adaptations additionally facilitate the determination of equilibrium binding metrics, making the assay a versatile tool for the study of receptor-ligand interactions.
Collapse
Affiliation(s)
| | - Martin Gustavsson
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark;
| |
Collapse
|
15
|
Atypical Roles of the Chemokine Receptor ACKR3/CXCR7 in Platelet Pathophysiology. Cells 2022; 11:cells11020213. [PMID: 35053329 PMCID: PMC8773869 DOI: 10.3390/cells11020213] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 12/23/2022] Open
Abstract
The manifold actions of the pro-inflammatory and regenerative chemokine CXCL12/SDF-1α are executed through the canonical GProteinCoupledReceptor CXCR4, and the non-canonical ACKR3/CXCR7. Platelets express CXCR4, ACKR3/CXCR7, and are a vital source of CXCL12/SDF-1α themselves. In recent years, a regulatory impact of the CXCL12-CXCR4-CXCR7 axis on platelet biogenesis, i.e., megakaryopoiesis, thrombotic and thrombo-inflammatory actions have been revealed through experimental and clinical studies. Platelet surface expression of ACKR3/CXCR7 is significantly enhanced following myocardial infarction (MI) in acute coronary syndrome (ACS) patients, and is also associated with improved functional recovery and prognosis. The therapeutic implications of ACKR3/CXCR7 in myocardial regeneration and improved recovery following an ischemic episode, are well documented. Cardiomyocytes, cardiac-fibroblasts, endothelial lining of the blood vessels perfusing the heart, besides infiltrating platelets and monocytes, all express ACKR3/CXCR7. This review recapitulates ligand induced differential trafficking of platelet CXCR4-ACKR3/CXCR7 affecting their surface availability, and in regulating thrombo-inflammatory platelet functions and survival through CXCR4 or ACKR3/CXCR7. It emphasizes the pro-thrombotic influence of CXCL12/SDF-1α exerted through CXCR4, as opposed to the anti-thrombotic impact of ACKR3/CXCR7. Offering an innovative translational perspective, this review also discusses the advantages and challenges of utilizing ACKR3/CXCR7 as a potential anti-thrombotic strategy in platelet-associated cardiovascular disorders, particularly in coronary artery disease (CAD) patients post-MI.
Collapse
|
16
|
Ehrlich AT, Semache M, Couvineau P, Wojcik S, Kobayashi H, Thelen M, Gross F, Hogue M, Le Gouill C, Darcq E, Bouvier M, Kieffer BL. Ackr3-Venus knock-in mouse lights up brain vasculature. Mol Brain 2021; 14:151. [PMID: 34583741 PMCID: PMC8477500 DOI: 10.1186/s13041-021-00862-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 09/17/2021] [Indexed: 01/09/2023] Open
Abstract
The atypical chemokine receptor 3, ACKR3, is a G protein-coupled receptor, which does not couple to G proteins but recruits βarrestins. At present, ACKR3 is considered a target for cancer and cardiovascular disorders, but less is known about the potential of ACKR3 as a target for brain disease. Further, mouse lines have been created to identify cells expressing the receptor, but there is no tool to visualize and study the receptor itself under physiological conditions. Here, we engineered a knock-in (KI) mouse expressing a functional ACKR3-Venus fusion protein to directly detect the receptor, particularly in the adult brain. In HEK-293 cells, native and fused receptors showed similar membrane expression, ligand induced trafficking and signaling profiles, indicating that the Venus fusion does not alter receptor signaling. We also found that ACKR3-Venus enables direct real-time monitoring of receptor trafficking using resonance energy transfer. In ACKR3-Venus knock-in mice, we found normal ACKR3 mRNA levels in the brain, suggesting intact gene transcription. We fully mapped receptor expression across 14 peripheral organs and 112 brain areas and found that ACKR3 is primarily localized to the vasculature in these tissues. In the periphery, receptor distribution aligns with previous reports. In the brain there is notable ACKR3 expression in endothelial vascular cells, hippocampal GABAergic interneurons and neuroblast neighboring cells. In conclusion, we have generated Ackr3-Venus knock-in mice with a traceable ACKR3 receptor, which will be a useful tool to the research community for interrogations about ACKR3 biology and related diseases.
Collapse
Affiliation(s)
- Aliza T Ehrlich
- Douglas Research Center, McGill University, Montréal, Canada.
- University of California, San Francisco, USA.
| | - Meriem Semache
- Institute for Research in Immunology and Cancer (IRIC) and Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, H3T 1J4, Canada
- Domain Therapeutics North America, Montréal, Québec, H4S 1Z9, Canada
| | - Pierre Couvineau
- Institute for Research in Immunology and Cancer (IRIC) and Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, H3T 1J4, Canada
| | - Stefan Wojcik
- Douglas Research Center, McGill University, Montréal, Canada
- University of Surrey, Guildford, UK
- Oxford Brookes University, Oxford, UK
| | - Hiroyuki Kobayashi
- Institute for Research in Immunology and Cancer (IRIC) and Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, H3T 1J4, Canada
| | - Marcus Thelen
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Florence Gross
- Institute for Research in Immunology and Cancer (IRIC) and Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, H3T 1J4, Canada
- Domain Therapeutics North America, Montréal, Québec, H4S 1Z9, Canada
| | - Mireille Hogue
- Institute for Research in Immunology and Cancer (IRIC) and Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, H3T 1J4, Canada
| | - Christian Le Gouill
- Institute for Research in Immunology and Cancer (IRIC) and Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, H3T 1J4, Canada
| | - Emmanuel Darcq
- Douglas Research Center, McGill University, Montréal, Canada
- INSERM U1114, University of Strasbourg, Strasbourg, France
| | - Michel Bouvier
- Institute for Research in Immunology and Cancer (IRIC) and Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, Québec, H3T 1J4, Canada.
| | - Brigitte L Kieffer
- Douglas Research Center, McGill University, Montréal, Canada.
- INSERM U1114, University of Strasbourg, Strasbourg, France.
| |
Collapse
|
17
|
Nucera F, Lo Bello F, Shen SS, Ruggeri P, Coppolino I, Di Stefano A, Stellato C, Casolaro V, Hansbro PM, Adcock IM, Caramori G. Role of Atypical Chemokines and Chemokine Receptors Pathways in the Pathogenesis of COPD. Curr Med Chem 2021; 28:2577-2653. [PMID: 32819230 DOI: 10.2174/0929867327999200819145327] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/11/2020] [Accepted: 06/18/2020] [Indexed: 11/22/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) represents a heightened inflammatory response in the lung generally resulting from tobacco smoking-induced recruitment and activation of inflammatory cells and/or activation of lower airway structural cells. Several mediators can modulate activation and recruitment of these cells, particularly those belonging to the chemokines (conventional and atypical) family. There is emerging evidence for complex roles of atypical chemokines and their receptors (such as high mobility group box 1 (HMGB1), antimicrobial peptides, receptor for advanced glycosylation end products (RAGE) or toll-like receptors (TLRs)) in the pathogenesis of COPD, both in the stable disease and during exacerbations. Modulators of these pathways represent potential novel therapies for COPD and many are now in preclinical development. Inhibition of only a single atypical chemokine or receptor may not block inflammatory processes because there is redundancy in this network. However, there are many animal studies that encourage studies for modulating the atypical chemokine network in COPD. Thus, few pharmaceutical companies maintain a significant interest in developing agents that target these molecules as potential antiinflammatory drugs. Antibody-based (biological) and small molecule drug (SMD)-based therapies targeting atypical chemokines and/or their receptors are mostly at the preclinical stage and their progression to clinical trials is eagerly awaited. These agents will most likely enhance our knowledge about the role of atypical chemokines in COPD pathophysiology and thereby improve COPD management.
Collapse
Affiliation(s)
- Francesco Nucera
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences (BIOMORF), University of Messina, Pugliatti Square 1, 98122 Messina, Italy
| | - Federica Lo Bello
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences (BIOMORF), University of Messina, Pugliatti Square 1, 98122 Messina, Italy
| | - Sj S Shen
- Faculty of Science, Centre for Inflammation, Centenary Institute, University of Technology, Ultimo, Sydney, Australia
| | - Paolo Ruggeri
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences (BIOMORF), University of Messina, Pugliatti Square 1, 98122 Messina, Italy
| | - Irene Coppolino
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences (BIOMORF), University of Messina, Pugliatti Square 1, 98122 Messina, Italy
| | - Antonino Di Stefano
- Division of Pneumology, Cyto- Immunopathology Laboratory of the Cardio-Respiratory System, Clinical Scientific Institutes Maugeri IRCCS, Veruno, Italy
| | - Cristiana Stellato
- Department of Medicine, Surgery and Dentistry, Salerno Medical School, University of Salerno, Salerno, Italy
| | - Vincenzo Casolaro
- Department of Medicine, Surgery and Dentistry, Salerno Medical School, University of Salerno, Salerno, Italy
| | - Phil M Hansbro
- Faculty of Science, Centre for Inflammation, Centenary Institute, University of Technology, Ultimo, Sydney, Australia
| | - Ian M Adcock
- Airway Disease Section, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Gaetano Caramori
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences (BIOMORF), University of Messina, Pugliatti Square 1, 98122 Messina, Italy
| |
Collapse
|
18
|
Differential Involvement of ACKR3 C-Tail in β-Arrestin Recruitment, Trafficking and Internalization. Cells 2021; 10:cells10030618. [PMID: 33799570 PMCID: PMC8002179 DOI: 10.3390/cells10030618] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
Background: The atypical chemokine receptor 3 (ACKR3) belongs to the superfamily of G protein-coupled receptors (GPCRs). Unlike classical GPCRs, this receptor does not activate G proteins in most cell types but recruits β-arrestins upon activation. ACKR3 plays an important role in cancer and vascular diseases. As recruitment of β-arrestins is triggered by phosphorylation of the C-terminal tail of GPCRs, we studied the role of different potential phosphorylation sites within the ACKR3 C-tail to further delineate the molecular mechanism of internalization and trafficking of this GPCR. Methods: We used various bioluminescence and fluorescence resonance energy transfer-based sensors and techniques in Human Embryonic Kidney (HEK) 293T cells expressing WT or phosphorylation site mutants of ACKR3 to measure CXCL12-induced recruitment of β-arrestins and G-protein-coupled receptor kinases (GRKs), receptor internalization and trafficking. Results: Upon CXCL12 stimulation, ACKR3 recruits both β-arrestin 1 and 2 with equivalent kinetic profiles. We identified interactions with GRK2, 3 and 5, with GRK2 and 3 being important for β-arrestin recruitment. Upon activation, ACKR3 internalizes and recycles back to the cell membrane. We demonstrate that β-arrestin recruitment to the receptor is mainly determined by a single cluster of phosphorylated residues on the C-tail of ACKR3, and that residue T352 and in part S355 are important residues for β-arrestin1 recruitment. Phosphorylation of the C-tail appears essential for ligand-induced internalization and important for differential β-arrestin recruitment. GRK2 and 3 play a key role in receptor internalization. Moreover, ACKR3 can still internalize when β-arrestin recruitment is impaired or in the absence of β-arrestins, using alternative internalization pathways. Our data indicate that distinct residues within the C-tail of ACKR3 differentially regulate CXCL12-induced β-arrestin recruitment, ACKR3 trafficking and internalization.
Collapse
|
19
|
Rusetska N, Kowalski K, Zalewski K, Zięba S, Bidziński M, Goryca K, Kotowicz B, Fuksiewicz M, Kopczynski J, Bakuła-Zalewska E, Kowalik A, Kowalewska M. CXCR4/ACKR3/CXCL12 axis in the lymphatic metastasis of vulvar squamous cell carcinoma. J Clin Pathol 2021; 75:324-332. [PMID: 33692092 PMCID: PMC9046756 DOI: 10.1136/jclinpath-2020-206917] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 01/05/2021] [Accepted: 01/24/2021] [Indexed: 12/15/2022]
Abstract
Aims Vulvar squamous cell carcinoma (VSCC) spreads early and mainly locally via direct expansion into adjacent structures, followed by lymphatic metastasis to the regional lymph nodes (LNs). In the lymphatic metastasis, cancer cells bearing CXCR4 and ACKR3 (CXCR7) receptors are recruited to the LNs that produce the CXCL12 ligand. Our study aimed to assess the role of the CXCR4/ACKR3/CXCL12 axis in VSCC progression. Methods Tumour and LN tissue samples were obtained from 46 patients with VSCC and 51 patients with premalignant vulvar lesions. We assessed CXCR4, ACKR3 and CXCL12 by immunohistochemistry (IHC) in the tissue samples. Additionally, CXCL12 levels were determined by ELISA in the sera of 23 patients with premalignant lesions, 37 with VSCC and 16 healthy volunteers. Results CXCR4 and ACKR3 proteins were virtually absent in vulvar precancers, while in VSCC samples the IHC staining was strong. In the LNs of patients with VSCC, 98% of metastatic cells expressed CXCR4 and 85% expressed ACKR3. Neither CXCR4 nor ACKR3 presence was correlated with tumour human papilloma virus status. Few CXCL12-positive cells were found in the analysed tissue samples, but serum CXCL12 levels were significantly increased in both patients with premalignant vulvar lesions and with VSCC compared with healthy volunteers. Conclusions It appears that during progression and lymphatic spread of VSCC, the CXCR4/ACKR3/CXCL12 axis is activated. Moreover, our data suggest that CXCR4 antagonists merit further attention as a possible therapeutic option in patients with VSCC.
Collapse
Affiliation(s)
- Natalia Rusetska
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Kamil Kowalski
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Kamil Zalewski
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland.,Department of Gynecologic Oncology, Holycross Cancer Center, Kielce, Poland.,Chair and Department of Obstetrics, Gynecology and Oncology, 2nd Faculty of Medicine, Warsaw Medical University, Warsaw, Poland
| | - Sebastian Zięba
- Department of Molecular Diagnostics, Holycross Cancer Center, Kielce, Poland
| | - Mariusz Bidziński
- Department of Gynecologic Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Krzysztof Goryca
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland.,Genomics Core Facility, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Beata Kotowicz
- Department of Pathology and Laboratory Diagnostics, Laboratory of Tumor Markers, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Malgorzata Fuksiewicz
- Department of Pathology and Laboratory Diagnostics, Laboratory of Tumor Markers, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Janusz Kopczynski
- Department of Surgical Pathology, Holycross Cancer Center, Kielce, Poland
| | - Elwira Bakuła-Zalewska
- Department of Pathology and Laboratory Diagnostics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Artur Kowalik
- Department of Molecular Diagnostics, Holycross Cancer Center, Kielce, Poland.,Division of Medical Biology, Jan Kochanowski University, Kielce, Poland
| | - Magdalena Kowalewska
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland .,Department of Immunology, Biochemistry and Nutrition, Centre for Preclinical Research and Technologies, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
20
|
Ligand-competent fractalkine receptor is expressed on exosomes. Biochem Biophys Rep 2021; 26:100932. [PMID: 33553692 PMCID: PMC7859287 DOI: 10.1016/j.bbrep.2021.100932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/18/2020] [Accepted: 01/11/2021] [Indexed: 11/28/2022] Open
Abstract
Expression of chemokine receptor CX3CR1 is reportedly restricted to several cell types including natural killer cells, cytotoxic T cells, monocytes, and macrophages. However, its expression and function on exosomes, which are nanosized extracellular vesicles known to act as mediators of intercellular communications, remain unclear. Here, we investigated CX3CR1 expression on exosomes isolated from various cell types. Although we found that all the exosomes tested in our study highly expressed CX3CR1, this chemokine receptor was expressed only inside, but barely on, their source cells. Moreover, exosomal CX3CR1 was capable of binding soluble CX3CL1. Therefore, our study suggests that CX3CR1 is a novel and ligand-competent exosome receptor. CX3CR1 is highly expressed by exosomes. Expression of CX3CR1 is restricted within, but not on, the cells. Exosomal CX3CR1 is capable of binding soluble CX3CL1. CX3CL1 binding of exosomes may deprive their source cells of the chance to bind this chemokine.
Collapse
|
21
|
Patwardhan A, Cheng N, Trejo J. Post-Translational Modifications of G Protein-Coupled Receptors Control Cellular Signaling Dynamics in Space and Time. Pharmacol Rev 2021; 73:120-151. [PMID: 33268549 PMCID: PMC7736832 DOI: 10.1124/pharmrev.120.000082] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are a large family comprising >800 signaling receptors that regulate numerous cellular and physiologic responses. GPCRs have been implicated in numerous diseases and represent the largest class of drug targets. Although advances in GPCR structure and pharmacology have improved drug discovery, the regulation of GPCR function by diverse post-translational modifications (PTMs) has received minimal attention. Over 200 PTMs are known to exist in mammalian cells, yet only a few have been reported for GPCRs. Early studies revealed phosphorylation as a major regulator of GPCR signaling, whereas later reports implicated a function for ubiquitination, glycosylation, and palmitoylation in GPCR biology. Although our knowledge of GPCR phosphorylation is extensive, our knowledge of the modifying enzymes, regulation, and function of other GPCR PTMs is limited. In this review we provide a comprehensive overview of GPCR post-translational modifications with a greater focus on new discoveries. We discuss the subcellular location and regulatory mechanisms that control post-translational modifications of GPCRs. The functional implications of newly discovered GPCR PTMs on receptor folding, biosynthesis, endocytic trafficking, dimerization, compartmentalized signaling, and biased signaling are also provided. Methods to detect and study GPCR PTMs as well as PTM crosstalk are further highlighted. Finally, we conclude with a discussion of the implications of GPCR PTMs in human disease and their importance for drug discovery. SIGNIFICANCE STATEMENT: Post-translational modification of G protein-coupled receptors (GPCRs) controls all aspects of receptor function; however, the detection and study of diverse types of GPCR modifications are limited. A thorough understanding of the role and mechanisms by which diverse post-translational modifications regulate GPCR signaling and trafficking is essential for understanding dysregulated mechanisms in disease and for improving and refining drug development for GPCRs.
Collapse
Affiliation(s)
- Anand Patwardhan
- Department of Pharmacology and the Biomedical Sciences Graduate Program, School of Medicine, University of California, San Diego, La Jolla, California
| | - Norton Cheng
- Department of Pharmacology and the Biomedical Sciences Graduate Program, School of Medicine, University of California, San Diego, La Jolla, California
| | - JoAnn Trejo
- Department of Pharmacology and the Biomedical Sciences Graduate Program, School of Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
22
|
Fumagalli A, Heuninck J, Pizzoccaro A, Moutin E, Koenen J, Séveno M, Durroux T, Junier MP, Schlecht-Louf G, Bachelerie F, Schütz D, Stumm R, Smit MJ, Guérineau NC, Chaumont-Dubel S, Marin P. The atypical chemokine receptor 3 interacts with Connexin 43 inhibiting astrocytic gap junctional intercellular communication. Nat Commun 2020; 11:4855. [PMID: 32978390 PMCID: PMC7519114 DOI: 10.1038/s41467-020-18634-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/02/2020] [Indexed: 12/12/2022] Open
Abstract
The atypical chemokine receptor 3 (ACKR3) plays a pivotal role in directing the migration of various cellular populations and its over-expression in tumors promotes cell proliferation and invasiveness. The intracellular signaling pathways transducing ACKR3-dependent effects remain poorly characterized, an issue we addressed by identifying the interactome of ACKR3. Here, we report that recombinant ACKR3 expressed in HEK293T cells recruits the gap junction protein Connexin 43 (Cx43). Cx43 and ACKR3 are co-expressed in mouse brain astrocytes and human glioblastoma cells and form a complex in embryonic mouse brain. Functional in vitro studies show enhanced ACKR3 interaction with Cx43 upon ACKR3 agonist stimulation. Furthermore, ACKR3 activation promotes β-arrestin2- and dynamin-dependent Cx43 internalization to inhibit gap junctional intercellular communication in primary astrocytes. These results demonstrate a functional link between ACKR3 and gap junctions that might be of pathophysiological relevance. The atypical chemokine receptor 3 (ACKR3) is known to regulate cell migration, but the underlying mechanisms are unclear. Here, the authors show, from an interactome analysis, ACKR3 association with the gap junction protein Connexin 43 in vivo and ACKR3-mediated inhibition of astrocyte gap junctional communication.
Collapse
Affiliation(s)
- Amos Fumagalli
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Joyce Heuninck
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Anne Pizzoccaro
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Enora Moutin
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Joyce Koenen
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, 92140, Clamart, France.,Amsterdam Institute for Molecules Medicines and Systems, Division of Medicinal Chemistry, Faculty of Sciences, VU University Amsterdam, 1081, HV, Amsterdam, The Netherlands
| | - Martial Séveno
- Biocampus Montpellier, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Thierry Durroux
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Marie-Pierre Junier
- CNRS UMR8246, Inserm U1130, Neuroscience Paris Seine-IBPS, Sorbonne Universités, Paris, France
| | - Géraldine Schlecht-Louf
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, 92140, Clamart, France
| | - Francoise Bachelerie
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, 92140, Clamart, France
| | - Dagmar Schütz
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747, Jena, Germany
| | - Ralf Stumm
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747, Jena, Germany
| | - Martine J Smit
- Amsterdam Institute for Molecules Medicines and Systems, Division of Medicinal Chemistry, Faculty of Sciences, VU University Amsterdam, 1081, HV, Amsterdam, The Netherlands
| | - Nathalie C Guérineau
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Séverine Chaumont-Dubel
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Philippe Marin
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France.
| |
Collapse
|
23
|
D'Agostino G, Artinger M, Locati M, Perez L, Legler DF, Bianchi ME, Rüegg C, Thelen M, Marchese A, Rocchi MBL, Cecchinato V, Uguccioni M. β-Arrestin1 and β-Arrestin2 Are Required to Support the Activity of the CXCL12/HMGB1 Heterocomplex on CXCR4. Front Immunol 2020; 11:550824. [PMID: 33072091 PMCID: PMC7533569 DOI: 10.3389/fimmu.2020.550824] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 08/21/2020] [Indexed: 12/14/2022] Open
Abstract
The chemokine receptor CXCR4 plays a fundamental role in homeostasis and pathology by orchestrating recruitment and positioning of immune cells, under the guidance of a CXCL12 gradient. The ability of chemokines to form heterocomplexes, enhancing their function, represents an additional level of regulation on their cognate receptors. In particular, the multi-faceted activity of the heterocomplex formed between CXCL12 and the alarmin HMGB1 is emerging as an unexpected player able to modulate a variety of cell responses, spanning from tissue regeneration to chronic inflammation. Nowadays, little is known on the selective signaling pathways activated when CXCR4 is triggered by the CXCL12/HMGB1 heterocomplex. In the present work, we demonstrate that this heterocomplex acts as a CXCR4 balanced agonist, activating both G protein and β-arrestins-mediated signaling pathways to sustain chemotaxis. We generated β-arrestins knock out HeLa cells by CRISPR/Cas9 technology and show that the CXCL12/HMGB1 heterocomplex-mediated actin polymerization is primarily β-arrestin1 dependent, while chemotaxis requires both β-arrestin1 and β-arrestin2. Triggering of CXCR4 with the CXCL12/HMGB1 heterocomplex leads to an unexpected receptor retention on the cell surface, which depends on β-arrestin2. In conclusion, the CXCL12/HMGB1 heterocomplex engages the β-arrestin proteins differently from CXCL12, promoting a prompt availability of CXCR4 on the cell surface, and enhancing directional cell migration. These data unveil the signaling induced by the CXCL12/HMGB1 heterocomplex in view of identifying biased CXCR4 antagonists or agonists targeting the variety of functions it exerts.
Collapse
Affiliation(s)
- Gianluca D'Agostino
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Marc Artinger
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, Kreuzlingen, Switzerland
| | - Massimo Locati
- Humanitas Clinical and Research Center IRCCS, Rozzano, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Laurent Perez
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Daniel F Legler
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, Kreuzlingen, Switzerland.,Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Marco E Bianchi
- Division of Genetics and Cell Biology, Vita-Salute San Raffaele University, Milan, Italy
| | - Curzio Rüegg
- Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Marcus Thelen
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Adriano Marchese
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Marco B L Rocchi
- Department of Biomolecular Sciences, Biostatistics Unit, University of Urbino, Urbino, Italy
| | - Valentina Cecchinato
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Mariagrazia Uguccioni
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| |
Collapse
|
24
|
Meyrath M, Szpakowska M, Zeiner J, Massotte L, Merz MP, Benkel T, Simon K, Ohnmacht J, Turner JD, Krüger R, Seutin V, Ollert M, Kostenis E, Chevigné A. The atypical chemokine receptor ACKR3/CXCR7 is a broad-spectrum scavenger for opioid peptides. Nat Commun 2020; 11:3033. [PMID: 32561830 PMCID: PMC7305236 DOI: 10.1038/s41467-020-16664-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 05/15/2020] [Indexed: 12/29/2022] Open
Abstract
Endogenous opioid peptides and prescription opioid drugs modulate pain, anxiety and stress by activating opioid receptors, currently classified into four subtypes. Here we demonstrate that ACKR3/CXCR7, hitherto known as an atypical scavenger receptor for chemokines, is a broad-spectrum scavenger of opioid peptides. Phylogenetically, ACKR3 is intermediate between chemokine and opioid receptors and is present in various brain regions together with classical opioid receptors. Functionally, ACKR3 is a scavenger receptor for a wide variety of opioid peptides, especially enkephalins and dynorphins, reducing their availability for the classical opioid receptors. ACKR3 is not modulated by prescription opioids, but we show that an ACKR3-selective subnanomolar competitor peptide, LIH383, can restrain ACKR3’s negative regulatory function on opioid peptides in rat brain and potentiate their activity towards classical receptors, which may open alternative therapeutic avenues for opioid-related disorders. Altogether, our results reveal that ACKR3 is an atypical opioid receptor with cross-family ligand selectivity. Opioids modulate pain, anxiety and stress by activating four subtypes of opioid receptors. The authors show that atypical chemokine receptor 3 (ACKR3) is a scavenger for various endogenous opioid peptides regulating their availability without activating downstream signaling.
Collapse
Affiliation(s)
- Max Meyrath
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), rue Henri Koch 29, L-4354, Esch-sur-Alzette, Luxembourg
| | - Martyna Szpakowska
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), rue Henri Koch 29, L-4354, Esch-sur-Alzette, Luxembourg
| | - Julian Zeiner
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115, Bonn, Germany
| | - Laurent Massotte
- Neurophysiology Unit, GIGA Neurosciences, University of Liège, avenue de l'hopital, B-4000, Liège, Belgium
| | - Myriam P Merz
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), rue Henri Koch 29, L-4354, Esch-sur-Alzette, Luxembourg
| | - Tobias Benkel
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115, Bonn, Germany.,Research Training Group 1873, University of Bonn, Bonn, Germany
| | - Katharina Simon
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115, Bonn, Germany
| | - Jochen Ohnmacht
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, avenue du Swing 6, L-4367, Belvaux, Luxembourg.,Department of Life Sciences and Medicine, University of Luxembourg, avenue du Swing 6, L-4367, Belvaux, Luxembourg
| | - Jonathan D Turner
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), rue Henri Koch 29, L-4354, Esch-sur-Alzette, Luxembourg
| | - Rejko Krüger
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, avenue du Swing 6, L-4367, Belvaux, Luxembourg.,Transversal Translational Medicine, Luxembourg Institute of Health (LIH), rue Thomas Edison 1A-B, L-1445, Strassen, Luxembourg
| | - Vincent Seutin
- Neurophysiology Unit, GIGA Neurosciences, University of Liège, avenue de l'hopital, B-4000, Liège, Belgium
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), rue Henri Koch 29, L-4354, Esch-sur-Alzette, Luxembourg.,Department of Dermatology and Allergy Center, Odense Research Center for Anaphylaxis, University of Southern Denmark, 5000, Odense, Denmark
| | - Evi Kostenis
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115, Bonn, Germany
| | - Andy Chevigné
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), rue Henri Koch 29, L-4354, Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
25
|
Lemos Duarte M, Devi LA. Post-translational Modifications of Opioid Receptors. Trends Neurosci 2020; 43:417-432. [PMID: 32459993 PMCID: PMC7323054 DOI: 10.1016/j.tins.2020.03.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/26/2020] [Accepted: 03/26/2020] [Indexed: 12/13/2022]
Abstract
Post-translational modifications (PTMs) are key events in signal transduction since they affect protein function by regulating their abundance and/or activity. PTMs involve the covalent attachment of functional groups to specific amino acids. Since they tend to be generally reversible, PTMs serve as regulators of signal transduction pathways. G-protein-coupled receptors (GPCRs) are major signaling proteins that undergo multiple types of PTMs. In this Review, we focus on the opioid receptors, members of GPCR family A, and highlight recent advances in the field that have underscored the importance of PTMs in the functional regulation of these receptors. Since opioid receptor activity plays a central role in the development of tolerance and addiction to morphine and other drugs of abuse, understanding the molecular mechanisms regulating receptor activity is of fundamental importance.
Collapse
Affiliation(s)
- Mariana Lemos Duarte
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lakshmi A Devi
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
26
|
Jaracz-Ros A, Bernadat G, Cutolo P, Gallego C, Gustavsson M, Cecon E, Baleux F, Kufareva I, Handel TM, Bachelerie F, Levoye A. Differential activity and selectivity of N-terminal modified CXCL12 chemokines at the CXCR4 and ACKR3 receptors. J Leukoc Biol 2020; 107:1123-1135. [PMID: 32374043 DOI: 10.1002/jlb.2ma0320-383rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 03/12/2020] [Accepted: 03/30/2020] [Indexed: 12/12/2022] Open
Abstract
Chemokines play critical roles in numerous physiologic and pathologic processes through their action on seven-transmembrane (TM) receptors. The N-terminal domain of chemokines, which is a key determinant of signaling via its binding within a pocket formed by receptors' TM helices, can be the target of proteolytic processing. An illustrative case of this regulatory mechanism is the natural processing of CXCL12 that generates chemokine variants lacking the first two N-terminal residues. Whereas such truncated variants behave as antagonists of CXCR4, the canonical G protein-coupled receptor of CXCL12, they are agonists of the atypical chemokine receptor 3 (ACKR3/CXCR7), suggesting the implication of different structural determinants in the complexes formed between CXCL12 and its two receptors. Recent analyses have suggested that the CXCL12 N-terminus first engages the TM helices of ACKR3 followed by the receptor N-terminus wrapping around the chemokine core. Here we investigated the first stage of ACKR3-CXCL12 interactions by comparing the activity of substituted or N-terminally truncated variants of CXCL12 toward CXCR4 and ACKR3. We showed that modification of the first two N-terminal residues of the chemokine (K1R or P2G) does not alter the ability of CXCL12 to activate ACKR3. Our results also identified the K1R variant as a G protein-biased agonist of CXCR4. Comparative molecular dynamics simulations of the complexes formed by ACKR3 either with CXCL12 or with the P2G variant identified interactions between the N-terminal 2-4 residues of CXCL12 and a pocket formed by receptor's TM helices 2, 6, and 7 as critical determinants for ACKR3 activation.
Collapse
Affiliation(s)
- Agnieszka Jaracz-Ros
- Université Paris-Saclay, INSERM, Inflammation, Microbiome and Immunosurveillance, Clamart, France
| | | | - Pasquale Cutolo
- Université Paris-Saclay, INSERM, Inflammation, Microbiome and Immunosurveillance, Clamart, France
| | - Carmen Gallego
- Université Paris-Saclay, INSERM, Inflammation, Microbiome and Immunosurveillance, Clamart, France
| | - Martin Gustavsson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, Jolla, Louisiana, California, USA
| | - Erika Cecon
- Université de Paris, Institut Cochin, CNRS, INSERM, Paris, France
| | - Françoise Baleux
- Institut Pasteur, Unité de Chimie des Biomolécules, Paris, France
| | - Irina Kufareva
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, Jolla, Louisiana, California, USA
| | - Tracy M Handel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, Jolla, Louisiana, California, USA
| | - Françoise Bachelerie
- Université Paris-Saclay, INSERM, Inflammation, Microbiome and Immunosurveillance, Clamart, France
| | - Angélique Levoye
- Université de Paris, PARCC, INSERM, Paris, France.,Université Sorbonne Paris Nord, Bobigny, France
| |
Collapse
|
27
|
Saaber F, Schütz D, Miess E, Abe P, Desikan S, Ashok Kumar P, Balk S, Huang K, Beaulieu JM, Schulz S, Stumm R. ACKR3 Regulation of Neuronal Migration Requires ACKR3 Phosphorylation, but Not β-Arrestin. Cell Rep 2020; 26:1473-1488.e9. [PMID: 30726732 DOI: 10.1016/j.celrep.2019.01.049] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 10/23/2018] [Accepted: 01/11/2019] [Indexed: 12/11/2022] Open
Abstract
Phosphorylation of heptahelical receptors is thought to regulate G protein signaling, receptor endocytosis, and non-canonical signaling via recruitment of β-arrestins. We investigated chemokine receptor functionality under phosphorylation-deficient and β-arrestin-deficient conditions by studying interneuron migration in the embryonic cortex. This process depends on CXCL12, CXCR4, G protein signaling and on the atypical CXCL12 receptor ACKR3. We found that phosphorylation was crucial, whereas β-arrestins were dispensable for ACKR3-mediated control of CXCL12 levels in vivo. Cortices of mice expressing phosphorylation-deficient ACKR3 exhibited a major interneuron migration defect, which was accompanied by excessive activation and loss of CXCR4. Cxcl12-overexpressing mice mimicked this phenotype. Excess CXCL12 caused lysosomal CXCR4 degradation, loss of CXCR4 responsiveness, and, ultimately, similar motility defects as Cxcl12 deficiency. By contrast, β-arrestin deficiency caused only a subtle migration defect mimicked by CXCR4 gain of function. These findings demonstrate that phosphorylation regulates atypical chemokine receptor function without β-arrestin involvement in chemokine sequestration and non-canonical signaling.
Collapse
Affiliation(s)
- Friederike Saaber
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany
| | - Dagmar Schütz
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany
| | - Elke Miess
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany
| | - Philipp Abe
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany
| | - Srinidhi Desikan
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany
| | - Praveen Ashok Kumar
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany
| | - Sara Balk
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany
| | - Ke Huang
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany
| | - Jean Martin Beaulieu
- Department of Pharmacology and Toxicology, University of Toronto, Toronto M5S 1A8, ON, Canada
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany
| | - Ralf Stumm
- Institute of Pharmacology and Toxicology, Jena University Hospital, 07747 Jena, Germany.
| |
Collapse
|
28
|
Lau S, Feitzinger A, Venkiteswaran G, Wang J, Lewellis SW, Koplinski CA, Peterson FC, Volkman BF, Meier-Schellersheim M, Knaut H. A negative-feedback loop maintains optimal chemokine concentrations for directional cell migration. Nat Cell Biol 2020; 22:266-273. [PMID: 32042179 PMCID: PMC7809593 DOI: 10.1038/s41556-020-0465-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 01/07/2020] [Indexed: 12/11/2022]
Abstract
Chemoattractant gradients frequently guide migrating cells. To achieve the most directional signal, such gradients should be maintained with concentrations around the dissociation constant (Kd)1-6 of the chemoreceptor. Whether this actually occurs in animals is unknown. Here we investigate whether a moving tissue, the zebrafish posterior lateral line primordium, buffers its attractant in this concentration range to achieve robust migration. We find that the Cxcl12 (also known as Sdf1) attractant gradient ranges from 0 to 12 nM, values similar to the 3.4 nM Kd of its receptor Cxcr4. When we increase the Kd of Cxcl12 for Cxcr4, primordium migration is less directional. Furthermore, a negative-feedback loop between Cxcl12 and its clearance receptor Ackr3 (also known as Cxcr7) regulates the Cxcl12 concentrations. Breaking this negative feedback by blocking the phosphorylation of the cytoplasmic tail of Ackr3 also results in less directional primordium migration. Thus, directed migration of the primordium is dependent on a close match between the Cxcl12 concentration and the Kd of Cxcl12 for Cxcr4, which is maintained by buffering of the chemokine levels. Quantitative modelling confirms the plausibility of this mechanism. We anticipate that buffering of attractant concentration is a general mechanism for ensuring robust cell migration.
Collapse
Affiliation(s)
- Stephanie Lau
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
| | - Anna Feitzinger
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
| | - Gayatri Venkiteswaran
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
| | - John Wang
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
| | - Stephen W Lewellis
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY, USA
| | - Chad A Koplinski
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Francis C Peterson
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Brian F Volkman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Martin Meier-Schellersheim
- Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Holger Knaut
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
29
|
Sjöberg E, Meyrath M, Chevigné A, Östman A, Augsten M, Szpakowska M. The diverse and complex roles of atypical chemokine receptors in cancer: From molecular biology to clinical relevance and therapy. Adv Cancer Res 2020; 145:99-138. [PMID: 32089166 DOI: 10.1016/bs.acr.2019.12.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Chemokines regulate directed cell migration, proliferation and survival and are key components in cancer biology. They exert their functions by interacting with seven-transmembrane domain receptors that signal through G proteins (GPCRs). A subgroup of four chemokine receptors known as the atypical chemokine receptors (ACKRs) has emerged as essential regulators of the chemokine functions. ACKRs play diverse and complex roles in tumor biology from tumor initiation to metastasis, including cancer cell proliferation, adherence to endothelium, epithelial-mesenchymal transition (EMT), extravasation from blood vessels, tumor-associated angiogenesis or protection from immunological responses. This chapter gives an overview on the established and emerging roles that the atypical chemokine receptors ACKR1, ACKR2, ACKR3 and ACKR4 play in the different phases of cancer development and dissemination, their clinical relevance, as well as on the hurdles to overcome in ACKRs targeting as cancer therapy.
Collapse
Affiliation(s)
- Elin Sjöberg
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Max Meyrath
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Andy Chevigné
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Arne Östman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | | | - Martyna Szpakowska
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
30
|
Wong M, Newton LR, Hartmann J, Hennrich ML, Wachsmuth M, Ronchi P, Guzmán-Herrera A, Schwab Y, Gavin AC, Gilmour D. Dynamic Buffering of Extracellular Chemokine by a Dedicated Scavenger Pathway Enables Robust Adaptation during Directed Tissue Migration. Dev Cell 2020; 52:492-508.e10. [PMID: 32059773 DOI: 10.1016/j.devcel.2020.01.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 11/22/2019] [Accepted: 01/13/2020] [Indexed: 01/16/2023]
Abstract
How tissues migrate robustly through changing guidance landscapes is poorly understood. Here, quantitative imaging is combined with inducible perturbation experiments to investigate the mechanisms that ensure robust tissue migration in vivo. We show that tissues exposed to acute "chemokine floods" halt transiently before they perfectly adapt, i.e., return to the baseline migration behavior in the continued presence of elevated chemokine levels. A chemokine-triggered phosphorylation of the atypical chemokine receptor Cxcr7b reroutes it from constitutive ubiquitination-regulated degradation to plasma membrane recycling, thus coupling scavenging capacity to extracellular chemokine levels. Finally, tissues expressing phosphorylation-deficient Cxcr7b migrate normally in the presence of physiological chemokine levels but show delayed recovery when challenged with elevated chemokine concentrations. This work establishes that adaptation to chemokine fluctuations can be "outsourced" from canonical GPCR signaling to an autonomously acting scavenger receptor that both senses and dynamically buffers chemokine levels to increase the robustness of tissue migration.
Collapse
Affiliation(s)
- Mie Wong
- Department of Molecular Life Sciences, University of Zürich, Winterthurerstrasse 190, 8057 Zurich, Switzerland; Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany.
| | - Lionel R Newton
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Jonas Hartmann
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Marco L Hennrich
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Malte Wachsmuth
- Luxendo GmbH, Kurfürsten-Anlage 58, 69115 Heidelberg, Germany
| | - Paolo Ronchi
- Electron Microscopy Core Facility, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Alejandra Guzmán-Herrera
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Yannick Schwab
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany; Electron Microscopy Core Facility, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany
| | - Anne-Claude Gavin
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany; Department for Cell Physiology and Metabolism, University of Geneva, 1 rue Michel Servet, 1211 Geneva 4, Switzerland
| | - Darren Gilmour
- Department of Molecular Life Sciences, University of Zürich, Winterthurerstrasse 190, 8057 Zurich, Switzerland; Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Meyerhofstraße 1, 69117 Heidelberg, Germany.
| |
Collapse
|
31
|
Lokeshwar BL, Kallifatidis G, Hoy JJ. Atypical chemokine receptors in tumor cell growth and metastasis. Adv Cancer Res 2020; 145:1-27. [PMID: 32089162 DOI: 10.1016/bs.acr.2019.12.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Atypical chemokine receptors (ACKRs) are seven-transmembrane cell surface protein receptors expressed in immune cells, normal mesenchymal cells, and several tumor cells. As of this writing, six ACKRs have been characterized by diverse activities. They bind both cysteine-cysteine (CC) type and cysteine-X-cysteine (CXC)-type chemokines, either alone, or together with a ligand bound-functional G-protein coupled (typical) chemokine receptor. The major structural difference between ACKRs and typical chemokine receptors is the substituted DRYLAIV amino acid motif in the second intracellular loop of the ACKR. Due to this substitution, these receptors cannot bind Gαi-type G-proteins responsible for intracellular calcium mobilization and cellular chemotaxis. Although initially characterized as non-signaling transmembrane receptors (decoy receptors) that attenuate ligand-induced signaling by GPCRs, studies of all ACKRs have shown ligand-independent and ligand-dependent transmembrane signaling in both non-tumor and tumor cells. The precise function and mechanism of the differential expression of ACKRs in many tumors are not understood well. The use of antagonists of ACKRs ligands has shown limited antitumor potential; however, depleting ACKR expression resulted in a reduction in experimental tumor growth and metastasis. The ACKRs represent a unique class of transmembrane signaling proteins that regulate growth, survival, and metastatic processes in tumor cells, affecting multiple pathways of tumor growth. Therefore, closer investigations of ACKRs have a high potential for identifying therapeutics which affect the intracellular signaling, preferentially via the ligand-independent mechanism.
Collapse
Affiliation(s)
- Bal L Lokeshwar
- Georgia Cancer Center, Augusta University, Augusta, GA, United States; Research Service, Charlie Norwood Veterans Administration Medical Center, Augusta, GA, United States.
| | - Georgios Kallifatidis
- Georgia Cancer Center, Augusta University, Augusta, GA, United States; Research Service, Charlie Norwood Veterans Administration Medical Center, Augusta, GA, United States; Department of Biological Sciences, Augusta University, Augusta, GA, United States
| | - James J Hoy
- LCMB Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| |
Collapse
|
32
|
Fumagalli A, Zarca A, Neves M, Caspar B, Hill SJ, Mayor F, Smit MJ, Marin P. CXCR4/ACKR3 Phosphorylation and Recruitment of Interacting Proteins: Key Mechanisms Regulating Their Functional Status. Mol Pharmacol 2019; 96:794-808. [PMID: 30837297 DOI: 10.1124/mol.118.115360] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 02/21/2019] [Indexed: 01/14/2023] Open
Abstract
The C-X-C motif chemokine receptor type 4 (CXCR4) and the atypical chemokine receptor 3 (ACKR3/CXCR7) are class A G protein-coupled receptors (GPCRs). Accumulating evidence indicates that GPCR subcellular localization, trafficking, transduction properties, and ultimately their pathophysiological functions are regulated by both interacting proteins and post-translational modifications. This has encouraged the development of novel techniques to characterize the GPCR interactome and to identify residues subjected to post-translational modifications, with a special focus on phosphorylation. This review first describes state-of-the-art methods for the identification of GPCR-interacting proteins and GPCR phosphorylated sites. In addition, we provide an overview of the current knowledge of CXCR4 and ACKR3 post-translational modifications and an exhaustive list of previously identified CXCR4- or ACKR3-interacting proteins. We then describe studies highlighting the importance of the reciprocal influence of CXCR4/ACKR3 interactomes and phosphorylation states. We also discuss their impact on the functional status of each receptor. These studies suggest that deeper knowledge of the CXCR4/ACKR3 interactomes along with their phosphorylation and ubiquitination status would shed new light on their regulation and pathophysiological functions.
Collapse
Affiliation(s)
- Amos Fumagalli
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Division of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (A.Z., M.J.S.); Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Madrid, Spain (M.N., F.M.); CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (M.N., F.M.); and Division of Physiology, Pharmacology and Neuroscience, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (B.C., S.J.H.)
| | - Aurélien Zarca
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Division of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (A.Z., M.J.S.); Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Madrid, Spain (M.N., F.M.); CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (M.N., F.M.); and Division of Physiology, Pharmacology and Neuroscience, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (B.C., S.J.H.)
| | - Maria Neves
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Division of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (A.Z., M.J.S.); Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Madrid, Spain (M.N., F.M.); CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (M.N., F.M.); and Division of Physiology, Pharmacology and Neuroscience, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (B.C., S.J.H.)
| | - Birgit Caspar
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Division of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (A.Z., M.J.S.); Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Madrid, Spain (M.N., F.M.); CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (M.N., F.M.); and Division of Physiology, Pharmacology and Neuroscience, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (B.C., S.J.H.)
| | - Stephen J Hill
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Division of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (A.Z., M.J.S.); Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Madrid, Spain (M.N., F.M.); CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (M.N., F.M.); and Division of Physiology, Pharmacology and Neuroscience, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (B.C., S.J.H.)
| | - Federico Mayor
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Division of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (A.Z., M.J.S.); Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Madrid, Spain (M.N., F.M.); CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (M.N., F.M.); and Division of Physiology, Pharmacology and Neuroscience, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (B.C., S.J.H.)
| | - Martine J Smit
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Division of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (A.Z., M.J.S.); Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Madrid, Spain (M.N., F.M.); CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (M.N., F.M.); and Division of Physiology, Pharmacology and Neuroscience, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (B.C., S.J.H.)
| | - Philippe Marin
- IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France (A.F., P.M.); Division of Medicinal Chemistry, Faculty of Science, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (A.Z., M.J.S.); Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Madrid, Spain (M.N., F.M.); CIBERCV, Instituto de Salud Carlos III, Madrid, Spain (M.N., F.M.); and Division of Physiology, Pharmacology and Neuroscience, Medical School, University of Nottingham, Queen's Medical Centre, Nottingham, United Kingdom (B.C., S.J.H.)
| |
Collapse
|
33
|
Heuninck J, Perpiñá Viciano C, Işbilir A, Caspar B, Capoferri D, Briddon SJ, Durroux T, Hill SJ, Lohse MJ, Milligan G, Pin JP, Hoffmann C. Context-Dependent Signaling of CXC Chemokine Receptor 4 and Atypical Chemokine Receptor 3. Mol Pharmacol 2019; 96:778-793. [PMID: 31092552 DOI: 10.1124/mol.118.115477] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 03/21/2019] [Indexed: 02/06/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are regulated by complex molecular mechanisms, both in physiologic and pathologic conditions, and their signaling can be intricate. Many factors influence their signaling behavior, including the type of ligand that activates the GPCR, the presence of interacting partners, the kinetics involved, or their location. The two CXC-type chemokine receptors, CXC chemokine receptor 4 (CXCR4) and atypical chemokine receptor 3 (ACKR3), both members of the GPCR superfamily, are important and established therapeutic targets in relation to cancer, human immunodeficiency virus infection, and inflammatory diseases. Therefore, it is crucial to understand how the signaling of these receptors works to be able to specifically target them. In this review, we discuss how the signaling pathways activated by CXCR4 and ACKR3 can vary in different situations. G protein signaling of CXCR4 depends on the cellular context, and discrepancies exist depending on the cell lines used. ACKR3, as an atypical chemokine receptor, is generally reported to not activate G proteins but can broaden its signaling spectrum upon heteromerization with other receptors, such as CXCR4, endothelial growth factor receptor, or the α 1-adrenergic receptor (α 1-AR). Also, CXCR4 forms heteromers with CC chemokine receptor (CCR) 2, CCR5, the Na+/H+ exchanger regulatory factor 1, CXCR3, α 1-AR, and the opioid receptors, which results in differential signaling from that of the monomeric subunits. In addition, CXCR4 is present on membrane rafts but can go into the nucleus during cancer progression, probably acquiring different signaling properties. In this review, we also provide an overview of the currently known critical amino acids involved in CXCR4 and ACKR3 signaling.
Collapse
Affiliation(s)
- Joyce Heuninck
- IGF, CNRS, Inserm, Université de Montpellier, Montpellier, France (J.H., T.D., J.-P.P.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.V., A.I., M.J.L., C.H.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (D.C., G.M.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (B.C., S.J.B., S.J.H.); and Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., S.J.B., S.J.H.)
| | - Cristina Perpiñá Viciano
- IGF, CNRS, Inserm, Université de Montpellier, Montpellier, France (J.H., T.D., J.-P.P.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.V., A.I., M.J.L., C.H.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (D.C., G.M.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (B.C., S.J.B., S.J.H.); and Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., S.J.B., S.J.H.)
| | - Ali Işbilir
- IGF, CNRS, Inserm, Université de Montpellier, Montpellier, France (J.H., T.D., J.-P.P.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.V., A.I., M.J.L., C.H.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (D.C., G.M.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (B.C., S.J.B., S.J.H.); and Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., S.J.B., S.J.H.)
| | - Birgit Caspar
- IGF, CNRS, Inserm, Université de Montpellier, Montpellier, France (J.H., T.D., J.-P.P.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.V., A.I., M.J.L., C.H.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (D.C., G.M.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (B.C., S.J.B., S.J.H.); and Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., S.J.B., S.J.H.)
| | - Davide Capoferri
- IGF, CNRS, Inserm, Université de Montpellier, Montpellier, France (J.H., T.D., J.-P.P.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.V., A.I., M.J.L., C.H.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (D.C., G.M.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (B.C., S.J.B., S.J.H.); and Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., S.J.B., S.J.H.)
| | - Stephen J Briddon
- IGF, CNRS, Inserm, Université de Montpellier, Montpellier, France (J.H., T.D., J.-P.P.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.V., A.I., M.J.L., C.H.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (D.C., G.M.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (B.C., S.J.B., S.J.H.); and Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., S.J.B., S.J.H.)
| | - Thierry Durroux
- IGF, CNRS, Inserm, Université de Montpellier, Montpellier, France (J.H., T.D., J.-P.P.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.V., A.I., M.J.L., C.H.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (D.C., G.M.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (B.C., S.J.B., S.J.H.); and Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., S.J.B., S.J.H.)
| | - Stephen J Hill
- IGF, CNRS, Inserm, Université de Montpellier, Montpellier, France (J.H., T.D., J.-P.P.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.V., A.I., M.J.L., C.H.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (D.C., G.M.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (B.C., S.J.B., S.J.H.); and Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., S.J.B., S.J.H.)
| | - Martin J Lohse
- IGF, CNRS, Inserm, Université de Montpellier, Montpellier, France (J.H., T.D., J.-P.P.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.V., A.I., M.J.L., C.H.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (D.C., G.M.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (B.C., S.J.B., S.J.H.); and Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., S.J.B., S.J.H.)
| | - Graeme Milligan
- IGF, CNRS, Inserm, Université de Montpellier, Montpellier, France (J.H., T.D., J.-P.P.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.V., A.I., M.J.L., C.H.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (D.C., G.M.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (B.C., S.J.B., S.J.H.); and Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., S.J.B., S.J.H.)
| | - Jean-Philippe Pin
- IGF, CNRS, Inserm, Université de Montpellier, Montpellier, France (J.H., T.D., J.-P.P.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.V., A.I., M.J.L., C.H.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (D.C., G.M.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (B.C., S.J.B., S.J.H.); and Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., S.J.B., S.J.H.)
| | - Carsten Hoffmann
- IGF, CNRS, Inserm, Université de Montpellier, Montpellier, France (J.H., T.D., J.-P.P.); Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.P.V., A.I., M.J.L., C.H.); Institute for Molecular Cell Biology, Centre for Molecular Biomedicine, University Hospital Jena, Friedrich Schiller University Jena, Jena, Germany (C.P.V., C.H.); Max Delbrück Center for Molecular Medicine, Berlin, Germany (A.I., M.J.L.); Centre for Translational Pharmacology, Institute of Molecular, Cell, and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom (D.C., G.M.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, United Kingdom (B.C., S.J.B., S.J.H.); and Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, United Kingdom (B.C., S.J.B., S.J.H.)
| |
Collapse
|
34
|
Quinn KE, Mackie DI, Caron KM. Emerging roles of atypical chemokine receptor 3 (ACKR3) in normal development and physiology. Cytokine 2019; 109:17-23. [PMID: 29903572 DOI: 10.1016/j.cyto.2018.02.024] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 02/21/2018] [Accepted: 02/21/2018] [Indexed: 01/16/2023]
Abstract
The discovery that atypical chemokine receptors (ACKRs) can initiate alternative signaling pathways rather than classical G-protein coupled receptor (GPCR) signaling has changed the paradigm of chemokine receptors and their roles in modulating chemotactic responses. The ACKR family has grown over the years, with discovery of new functions and roles in a variety of pathophysiological conditions. However, the extent to which these receptors regulate normal physiology is still continuously expanding. In particular, atypical chemokine receptor 3 (ACKR3) has proven to be an important receptor in mediating normal biological functions, including cardiac development and migration of cortical neurons. In this review, we illustrate the versatile and intriguing role of ACKR3 in physiology.
Collapse
Affiliation(s)
- K E Quinn
- Department of Cell Biology and Physiology, 111 MasonFarm Rd., 6312B MBRB CB# 7545, The University of North Carolina, Chapel Hill, NC 27599-7545, USA
| | - D I Mackie
- Department of Cell Biology and Physiology, 111 MasonFarm Rd., 6312B MBRB CB# 7545, The University of North Carolina, Chapel Hill, NC 27599-7545, USA
| | - K M Caron
- Department of Cell Biology and Physiology, 111 MasonFarm Rd., 6312B MBRB CB# 7545, The University of North Carolina, Chapel Hill, NC 27599-7545, USA.
| |
Collapse
|
35
|
Gustavsson M, Dyer DP, Zhao C, Handel TM. Kinetics of CXCL12 binding to atypical chemokine receptor 3 reveal a role for the receptor N terminus in chemokine binding. Sci Signal 2019; 12:12/598/eaaw3657. [PMID: 31506383 DOI: 10.1126/scisignal.aaw3657] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Chemokines bind to membrane-spanning chemokine receptors, which signal through G proteins and promote cell migration. However, atypical chemokine receptor 3 (ACKR3) does not appear to couple to G proteins, and instead of directly promoting cell migration, it regulates the extracellular concentration of chemokines that it shares with the G protein-coupled receptors (GPCRs) CXCR3 and CXCR4, thereby influencing the responses of these receptors. Understanding how these receptors bind their ligands is important for understanding these different processes. Here, we applied association and dissociation kinetic measurements coupled to β-arrestin recruitment assays to investigate ACKR3:chemokine interactions. Our results showed that CXCL12 binding is unusually slow and driven by the interplay between multiple binding epitopes. We also found that the amino terminus of the receptor played a key role in chemokine binding and activation by preventing chemokine dissociation. It was thought that chemokines initially bind receptors through interactions between the globular domain of the chemokine and the receptor amino terminus, which then guides the chemokine amino terminus into the transmembrane pocket of the receptor to initiate signaling. On the basis of our kinetic data, we propose an alternative mechanism in which the amino terminus of the chemokine initially forms interactions with the extracellular loops and transmembrane pocket of the receptor, which is followed by the receptor amino terminus wrapping around the core of the chemokine to prolong its residence time. These data provide insight into how ACKR3 competes and cooperates with canonical GPCRs in its function as a scavenger receptor.
Collapse
Affiliation(s)
- Martin Gustavsson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0684, USA
| | - Douglas P Dyer
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0684, USA
| | - Chunxia Zhao
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0684, USA
| | - Tracy M Handel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0684, USA.
| |
Collapse
|
36
|
Bai Y, Yang Y, Yan Y, Zhong J, Blee AM, Pan Y, Ma T, Karnes RJ, Jimenez R, Xu W, Huang H. RUNX2 overexpression and PTEN haploinsufficiency cooperate to promote CXCR7 expression and cellular trafficking, AKT hyperactivation and prostate tumorigenesis. Theranostics 2019; 9:3459-3475. [PMID: 31281490 PMCID: PMC6587168 DOI: 10.7150/thno.33292] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 05/06/2019] [Indexed: 12/12/2022] Open
Abstract
Rationale: The overall success rate of prostate cancer (PCa) diagnosis and therapy has been improved over the years. However, genomic and phenotypic heterogeneity remains a major challenge for effective detection and treatment of PCa. Efforts to better classify PCa into functional subtypes and elucidate the molecular mechanisms underlying prostate tumorigenesis and therapy resistance are warranted for further improvement of PCa outcomes. Methods: We generated Cre+;Runx2-cTg;Ptenp/+ (Runx2-Pten double mutant) mice by crossbreeding Cre+;Runx2-cTg males with Pten conditional (Ptenp/p) females. By using Hematoxylin and Eosin (H&E) staining, SMA and Masson's Trichrome staining, we investigated the effect of PTEN haploinsufficiency in combination with Runx2 overexpression on prostate tumorigenesis. Moreover, we employed immunohistochemistry (IHC) to stain Ki67 for cell proliferation, cleaved caspase 3 for apoptosis and AKT phosphorylation for signaling pathway in prostate tissues. Chromatin immunoprecipitation coupled quantitative PCR (ChIP-qPCR), reverse transcription coupled quantitative PCR (RT-qPCR), western blot (WB) analyses and immunofluorescence (IF) were conducted to determine the underlying mechanism by which RUNX2 regulates CXCR7 and AKT phosphorylation in PCa cells. Results: We demonstrated that mice with prostate-specific Pten heterozygous deletion and Runx2 overexpression developed high-grade prostatic intraepithelial neoplasia (HGPIN) and cancerous lesions at age younger than one year, with concomitant high level expression of Akt phosphorylation and the chemokine receptor Cxcr7 in malignant glands. RUNX2 overexpression induced CXCR7 transcription and membrane location and AKT phosphorylation in PTEN-deficient human PCa cell lines. Increased expression of RUNX2 also promoted growth of PCa cells and this effect was largely mediated by CXCR7. CXCR7 expression also positively correlated with AKT phosphorylation in PCa patient specimens. Conclusions: Our results reveal a previously unidentified cooperative role of RUNX2 overexpression and PTEN haploinsufficiency in prostate tumorigenesis, suggesting that the defined RUNX2-CXCR7-AKT axis can be a viable target for effective treatment of PCa.
Collapse
Affiliation(s)
- Yang Bai
- Heilongjiang Key Laboratory of Scientific Research in Urology and Department of Urology, the Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150081, China
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Yinhui Yang
- Heilongjiang Key Laboratory of Scientific Research in Urology and Department of Urology, the Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150081, China
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Yuqian Yan
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Jian Zhong
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Alexandra M. Blee
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Yunqian Pan
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Tao Ma
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - R. Jeffrey Karnes
- Department of Urology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Rafael Jimenez
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Wanhai Xu
- Heilongjiang Key Laboratory of Scientific Research in Urology and Department of Urology, the Fourth Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150081, China
| | - Haojie Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
- Department of Urology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
- Mayo Clinic Cancer Center, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| |
Collapse
|
37
|
Borroni EM, Savino B, Bonecchi R, Locati M. Chemokines sound the alarmin: The role of atypical chemokine in inflammation and cancer. Semin Immunol 2018; 38:63-71. [DOI: 10.1016/j.smim.2018.10.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 10/08/2018] [Indexed: 12/17/2022]
|
38
|
Ulloa-Aguirre A, Zariñán T, Gutiérrez-Sagal R, Dias JA. Intracellular Trafficking of Gonadotropin Receptors in Health and Disease. Handb Exp Pharmacol 2018; 245:1-39. [PMID: 29063275 DOI: 10.1007/164_2017_49] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Gonadotropin receptors belong to the highly conserved subfamily of the G protein-coupled receptor (GPCR) superfamily, the so-called Rhodopsin-like family (class A), which is the largest class of GPCRs and currently a major drug target. Both the follicle-stimulating hormone receptor (FSHR) and the luteinizing hormone/chorionic gonadotropin hormone receptor (LHCGR) are mainly located in the gonads where they play key functions associated to essential reproductive functions. As any other protein, gonadotropin receptors must be properly folded into a mature tertiary conformation compatible with quaternary assembly and endoplasmic reticulum export to the cell surface plasma membrane. Several primary and secondary structural features, including presence of particular amino acid residues and short motifs and in addition, posttranslational modifications, regulate intracellular trafficking of gonadotropin receptors to the plasma membrane as well as internalization and recycling of the receptor back to the cell surface after activation by agonist. Inactivating mutations of gonadotropin receptors may derive from receptor misfolding and lead to absent or reduced plasma membrane expression of the altered receptor, thereby manifesting an array of phenotypical abnormalities mostly characterized by reproductive failure and/or abnormal or absence of development of secondary sex characteristics. In this chapter we review the structural requirements necessary for intracellular trafficking of the gonadotropin receptors, and describe how mutations in these receptors may lead to receptor misfolding and disease in humans.
Collapse
Affiliation(s)
- Alfredo Ulloa-Aguirre
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México-Instituto Nacional de Ciencias Médicas y Nutrición SZ, Vasco de Quiroga 15, Tlalpan, Mexico City, 14000, Mexico.
| | - Teresa Zariñán
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México-Instituto Nacional de Ciencias Médicas y Nutrición SZ, Vasco de Quiroga 15, Tlalpan, Mexico City, 14000, Mexico
| | - Rubén Gutiérrez-Sagal
- Red de Apoyo a la Investigación (RAI), Universidad Nacional Autónoma de México-Instituto Nacional de Ciencias Médicas y Nutrición SZ, Vasco de Quiroga 15, Tlalpan, Mexico City, 14000, Mexico
| | - James A Dias
- Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, NY, USA
| |
Collapse
|
39
|
Montpas N, St-Onge G, Nama N, Rhainds D, Benredjem B, Girard M, Hickson G, Pons V, Heveker N. Ligand-specific conformational transitions and intracellular transport are required for atypical chemokine receptor 3-mediated chemokine scavenging. J Biol Chem 2017; 293:893-905. [PMID: 29180449 PMCID: PMC5777261 DOI: 10.1074/jbc.m117.814947] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 11/20/2017] [Indexed: 11/06/2022] Open
Abstract
The atypical chemokine receptor ACKR3 contributes to chemotaxis by binding, internalizing, and degrading the chemokines CXCL11 and CXCL12 to shape and terminate chemotactic gradients during development and immune responses. Although unable to trigger G protein activation, both ligands activate G protein-independent ACKR3 responses and prompt arrestin recruitment. This offers a model to specifically study ligand-specific receptor conformations leading to G protein-independent signaling and to functional parameters such as receptor transport and chemokine degradation. We here show chemokine specificity in arrestin recruitment, by different effects of single amino acid substitutions in ACKR3 on arrestin in response to CXCL12 or CXCL11. Chemokine specificity in receptor transport was also observed, as CXCL11 induced faster receptor internalization, slower recycling, and longer intracellular sojourn of ACKR3 than CXCL12. Internalization and recycling rates of the ACKR3 R1423.50A substitution in response to each chemokine were similar; however, ACKR3 R1423.50A degraded only CXCL12 and not CXCL11. This suggests that ligand-specific intracellular receptor transport is required for chemokine degradation. Remarkably, the failure of ACKR3 R1423.50A to degrade CXCL11 was not caused by the lack of arrestin recruitment; rather, arrestin was entirely dispensable for scavenging of either chemokine. This suggests the involvement of another, yet unidentified, ACKR3 effector in scavenging. In summary, our study correlates ACKR3 ligand-specific conformational transitions with chemokine-dependent receptor transport dynamics and points toward unexpected ligand specificity in the mechanisms of chemokine degradation.
Collapse
Affiliation(s)
- Nicolas Montpas
- From the Department of Biochemistry and Molecular Medicine, University of Montréal, Montréal, Quebec H3T 1J4, Canada.,the Research Centre, Saint-Justine Hospital, University of Montréal, Montréal, Quebec H3T 1C5, Canada
| | - Geneviève St-Onge
- the Research Centre, Saint-Justine Hospital, University of Montréal, Montréal, Quebec H3T 1C5, Canada
| | - Nassr Nama
- From the Department of Biochemistry and Molecular Medicine, University of Montréal, Montréal, Quebec H3T 1J4, Canada.,the Research Centre, Saint-Justine Hospital, University of Montréal, Montréal, Quebec H3T 1C5, Canada
| | - David Rhainds
- From the Department of Biochemistry and Molecular Medicine, University of Montréal, Montréal, Quebec H3T 1J4, Canada.,the Research Centre, Saint-Justine Hospital, University of Montréal, Montréal, Quebec H3T 1C5, Canada
| | - Besma Benredjem
- From the Department of Biochemistry and Molecular Medicine, University of Montréal, Montréal, Quebec H3T 1J4, Canada.,the Research Centre, Saint-Justine Hospital, University of Montréal, Montréal, Quebec H3T 1C5, Canada
| | - Mélanie Girard
- From the Department of Biochemistry and Molecular Medicine, University of Montréal, Montréal, Quebec H3T 1J4, Canada.,the Research Centre, Saint-Justine Hospital, University of Montréal, Montréal, Quebec H3T 1C5, Canada
| | - Gilles Hickson
- the Research Centre, Saint-Justine Hospital, University of Montréal, Montréal, Quebec H3T 1C5, Canada.,the Department of Pathology and Cell Biology, University of Montréal, Montréal, Quebec H3T 1J4, Canada, and
| | - Véronique Pons
- INSERM, UMR 1048, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, F-31432 Toulouse, France
| | - Nikolaus Heveker
- From the Department of Biochemistry and Molecular Medicine, University of Montréal, Montréal, Quebec H3T 1J4, Canada, .,the Research Centre, Saint-Justine Hospital, University of Montréal, Montréal, Quebec H3T 1C5, Canada
| |
Collapse
|
40
|
Regulation of G Protein-Coupled Receptors by Ubiquitination. Int J Mol Sci 2017; 18:ijms18050923. [PMID: 28448471 PMCID: PMC5454836 DOI: 10.3390/ijms18050923] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 04/20/2017] [Accepted: 04/23/2017] [Indexed: 02/07/2023] Open
Abstract
G protein-coupled receptors (GPCRs) comprise the largest family of membrane receptors that control many cellular processes and consequently often serve as drug targets. These receptors undergo a strict regulation by mechanisms such as internalization and desensitization, which are strongly influenced by posttranslational modifications. Ubiquitination is a posttranslational modification with a broad range of functions that is currently gaining increased appreciation as a regulator of GPCR activity. The role of ubiquitination in directing GPCRs for lysosomal degradation has already been well-established. Furthermore, this modification can also play a role in targeting membrane and endoplasmic reticulum-associated receptors to the proteasome. Most recently, ubiquitination was also shown to be involved in GPCR signaling. In this review, we present current knowledge on the molecular basis of GPCR regulation by ubiquitination, and highlight the importance of E3 ubiquitin ligases, deubiquitinating enzymes and β-arrestins. Finally, we discuss classical and newly-discovered functions of ubiquitination in controlling GPCR activity.
Collapse
|
41
|
Stone MJ, Hayward JA, Huang C, E Huma Z, Sanchez J. Mechanisms of Regulation of the Chemokine-Receptor Network. Int J Mol Sci 2017; 18:E342. [PMID: 28178200 PMCID: PMC5343877 DOI: 10.3390/ijms18020342] [Citation(s) in RCA: 196] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 01/18/2017] [Accepted: 01/26/2017] [Indexed: 12/18/2022] Open
Abstract
The interactions of chemokines with their G protein-coupled receptors promote the migration of leukocytes during normal immune function and as a key aspect of the inflammatory response to tissue injury or infection. This review summarizes the major cellular and biochemical mechanisms by which the interactions of chemokines with chemokine receptors are regulated, including: selective and competitive binding interactions; genetic polymorphisms; mRNA splice variation; variation of expression, degradation and localization; down-regulation by atypical (decoy) receptors; interactions with cell-surface glycosaminoglycans; post-translational modifications; oligomerization; alternative signaling responses; and binding to natural or pharmacological inhibitors.
Collapse
Affiliation(s)
- Martin J Stone
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia.
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia.
| | - Jenni A Hayward
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia.
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia.
| | - Cheng Huang
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia.
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia.
| | - Zil E Huma
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia.
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia.
| | - Julie Sanchez
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia.
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
42
|
de Munnik SM, van der Lee R, Velders DM, van Offenbeek J, Smits-de Vries L, Leurs R, Smit MJ, Vischer HF. The viral G protein-coupled receptor ORF74 unmasks phospholipase C signaling of the receptor tyrosine kinase IGF-1R. Cell Signal 2016; 28:595-605. [DOI: 10.1016/j.cellsig.2016.02.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 02/25/2016] [Accepted: 02/25/2016] [Indexed: 11/16/2022]
|
43
|
Massara M, Bonavita O, Mantovani A, Locati M, Bonecchi R. Atypical chemokine receptors in cancer: friends or foes? J Leukoc Biol 2016; 99:927-33. [PMID: 26908826 DOI: 10.1189/jlb.3mr0915-431rr] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 02/02/2016] [Indexed: 01/23/2023] Open
Abstract
The chemokine system is a fundamental component of cancer-related inflammation involved in all stages of cancer development. It controls not only leukocyte infiltration in primary tumors but also angiogenesis, cancer cell proliferation, and migration to metastatic sites. Atypical chemokine receptors are a new, emerging class of regulators of the chemokine system. They control chemokine bioavailability by scavenging, transporting, or storing chemokines. They can also regulate the activity of canonical chemokine receptors with which they share the ligands by forming heterodimers or by modulating their expression levels or signaling activity. Here, we summarize recent results about the role of these receptors (atypical chemokine receptor 1/Duffy antigen receptor for chemokine, atypical chemokine receptor 2/D6, atypical chemokine receptor 3/CXC-chemokine receptor 7, and atypical chemokine receptor 4/CC-chemokine receptor-like 1) on the tumorigenesis process, indicating that their effects are strictly dependent on the cell type on which they are expressed and on their coexpression with other chemokine receptors. Indeed, atypical chemokine receptors inhibit tumor growth and progression through their activity as negative regulators of chemokine bioavailability, whereas, on the contrary, they can promote tumorigenesis when they regulate the signaling of other chemokine receptors, such as CXC-chemokine receptor 4. Thus, atypical chemokine receptors are key components of the regulatory network of inflammation and immunity in cancer and may have a major effect on anti-inflammatory and immunotherapeutic strategies.
Collapse
Affiliation(s)
- Matteo Massara
- Humanitas Clinical and Research Center, Rozzano, Italy; Department of Medical Biotechnologies and Translational Medicine, Università degli Studi di Milano, Rozzano, Italy; and
| | - Ornella Bonavita
- Humanitas Clinical and Research Center, Rozzano, Italy; Department of Medical Biotechnologies and Translational Medicine, Università degli Studi di Milano, Rozzano, Italy; and
| | - Alberto Mantovani
- Humanitas Clinical and Research Center, Rozzano, Italy; Department of Biomedical Sciences, Humanitas University, Rozzano, Italy
| | - Massimo Locati
- Humanitas Clinical and Research Center, Rozzano, Italy; Department of Medical Biotechnologies and Translational Medicine, Università degli Studi di Milano, Rozzano, Italy; and
| | - Raffaella Bonecchi
- Humanitas Clinical and Research Center, Rozzano, Italy; Department of Biomedical Sciences, Humanitas University, Rozzano, Italy
| |
Collapse
|
44
|
Milanos L, Brox R, Frank T, Poklukar G, Palmisano R, Waibel R, Einsiedel J, Dürr M, Ivanović-Burmazović I, Larsen O, Hjortø GM, Rosenkilde MM, Tschammer N. Discovery and Characterization of Biased Allosteric Agonists of the Chemokine Receptor CXCR3. J Med Chem 2016; 59:2222-43. [DOI: 10.1021/acs.jmedchem.5b01965] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Lampros Milanos
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| | - Regine Brox
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| | - Theresa Frank
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| | - Gašper Poklukar
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
- Faculty
of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Ralf Palmisano
- Optical
Imaging Center Erlangen, Friedrich Alexander University, Hartmannstraße
14, 91052 Erlangen, Germany
| | - Reiner Waibel
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| | - Jürgen Einsiedel
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| | - Maximilian Dürr
- Department
of Chemistry and Pharmacy, Bioorganic Chemistry, Friedrich Alexander University, Egerlandstraße 1, 91058 Erlangen, Germany
| | - Ivana Ivanović-Burmazović
- Department
of Chemistry and Pharmacy, Bioorganic Chemistry, Friedrich Alexander University, Egerlandstraße 1, 91058 Erlangen, Germany
| | - Olav Larsen
- Department
of Neuroscience and Pharmacology, Laboratory for Molecular Pharmacology,
Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen N, Denmark
| | - Gertrud Malene Hjortø
- Department
of Neuroscience and Pharmacology, Laboratory for Molecular Pharmacology,
Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen N, Denmark
| | - Mette Marie Rosenkilde
- Department
of Neuroscience and Pharmacology, Laboratory for Molecular Pharmacology,
Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen N, Denmark
| | - Nuska Tschammer
- Department
of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University, Schuhstraße 19, 91052 Erlangen, Germany
| |
Collapse
|
45
|
Vacchini A, Locati M, Borroni EM. Overview and potential unifying themes of the atypical chemokine receptor family. J Leukoc Biol 2016; 99:883-92. [PMID: 26740381 DOI: 10.1189/jlb.2mr1015-477r] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 12/12/2015] [Indexed: 12/17/2022] Open
Abstract
Chemokines modulate immune responses through their ability to orchestrate the migration of target cells. Chemokines directly induce cell migration through a distinct set of 7 transmembrane domain G protein-coupled receptors but are also recognized by a small subfamily of atypical chemokine receptors, characterized by their inability to support chemotactic activity. Atypical chemokine receptors are now emerging as crucial regulatory components of chemokine networks in a wide range of physiologic and pathologic contexts. Although a new nomenclature has been approved recently to reflect their functional distinction from their conventional counterparts, a systematic view of this subfamily is still missing. This review discusses their biochemical and immunologic properties to identify potential unifying themes in this emerging family.
Collapse
Affiliation(s)
- Alessandro Vacchini
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, and Humanitas Clinical and Research Center, Milan, Italy
| | - Massimo Locati
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, and Humanitas Clinical and Research Center, Milan, Italy
| | - Elena Monica Borroni
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, and Humanitas Clinical and Research Center, Milan, Italy
| |
Collapse
|
46
|
Jean-Charles PY, Snyder JC, Shenoy SK. Chapter One - Ubiquitination and Deubiquitination of G Protein-Coupled Receptors. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 141:1-55. [PMID: 27378754 DOI: 10.1016/bs.pmbts.2016.05.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The seven-transmembrane containing G protein-coupled receptors (GPCRs) constitute the largest family of cell-surface receptors. Transmembrane signaling by GPCRs is fundamental to many aspects of physiology including vision, olfaction, cardiovascular, and reproductive functions as well as pain, behavior and psychomotor responses. The duration and magnitude of signal transduction is tightly controlled by a series of coordinated trafficking events that regulate the cell-surface expression of GPCRs at the plasma membrane. Moreover, the intracellular trafficking profiles of GPCRs can correlate with the signaling efficacy and efficiency triggered by the extracellular stimuli that activate GPCRs. Of the various molecular mechanisms that impart selectivity, sensitivity and strength of transmembrane signaling, ubiquitination of the receptor protein plays an important role because it defines both trafficking and signaling properties of the activated GPCR. Ubiquitination of proteins was originally discovered in the context of lysosome-independent degradation of cytosolic proteins by the 26S proteasome; however a large body of work suggests that ubiquitination also orchestrates the downregulation of membrane proteins in the lysosomes. In the case of GPCRs, such ubiquitin-mediated lysosomal degradation engenders long-term desensitization of transmembrane signaling. To date about 40 GPCRs are known to be ubiquitinated. For many GPCRs, ubiquitination plays a major role in postendocytic trafficking and sorting to the lysosomes. This chapter will focus on the patterns and functional roles of GPCR ubiquitination, and will describe various molecular mechanisms involved in GPCR ubiquitination.
Collapse
Affiliation(s)
- P-Y Jean-Charles
- Department of Medicine (Cardiology), Duke University Medical Center, Durham, NC, United States
| | - J C Snyder
- Department of Cell Biology, Duke University Medical Center, Durham, NC, United States
| | - S K Shenoy
- Department of Medicine (Cardiology), Duke University Medical Center, Durham, NC, United States; Department of Cell Biology, Duke University Medical Center, Durham, NC, United States.
| |
Collapse
|
47
|
Wang L, Li X, Zhao Y, Fang C, Lian Y, Gou W, Han T, Zhu X. Insights into the mechanism of CXCL12-mediated signaling in trophoblast functions and placental angiogenesis. Acta Biochim Biophys Sin (Shanghai) 2015; 47:663-72. [PMID: 26188201 DOI: 10.1093/abbs/gmv064] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 06/08/2015] [Indexed: 01/07/2023] Open
Abstract
The chemokine CXCL12 and its receptor CXCR4 are important signaling components required for human blastocyst implantation and the progression of pregnancy. Growing evidence indicates that the CXCL12/CXCR4 axis can regulate trophoblast function and uterine spiral artery remodeling, which plays a fundamental role in placentation and fetal outcome. The orphan receptor CXCR7 is also believed to partly regulate the function of the CXCL12/CXCR4 axis. Additionally, the CXCL12/CXCR4/CXCR7 axis can enhance the cross-talk between trophoblasts and decidual cells such as uterine natural killer cells and decidual stromal cells which are involved in regulation of trophoblast differentiation and invasion and placental angiogenesis. In addition, recent studies proved that CXCL12 expression is elevated in the placenta and mid-trimester amniotic fluid of pregnant women with preeclampsia, implying that dysregulation of CXCL12 plays a role in the pathogenesis of preeclampsia. Further understanding of the regulatory mechanisms of CXCL12-mediated signaling in trophoblast functions and placental angiogenesis may help to design novel therapeutic approaches for pregnancy-associated diseases.
Collapse
Affiliation(s)
- Liang Wang
- Department of Obstetrics and Gynecology, Tangdu Hospital, the Fourth Military Medical University, Xi'an 710038, China The First Student Brigade, The Fourth Military Medical University, Xi'an 710032, China
| | - Xueyi Li
- Department of Clinical Immunology, PLA Specialized Research Institute of Rheumatism & Immunity, Xi-jing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Yilin Zhao
- Department of Respiratory Medicine, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, China
| | - Chao Fang
- Institute of Neurosciences, School of Basic Medical Sciences, The Fourth Military Medical University, Xi'an 710032, China
| | - Yingli Lian
- Department of Obstetrics and Gynecology, Tangdu Hospital, the Fourth Military Medical University, Xi'an 710038, China
| | - Wenli Gou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Tao Han
- Department of Orthopedics, Hainan Branch of PLA General Hospital, Sanya 572013, China
| | - Xiaoming Zhu
- Department of Obstetrics and Gynecology, Tangdu Hospital, the Fourth Military Medical University, Xi'an 710038, China
| |
Collapse
|
48
|
Pawig L, Klasen C, Weber C, Bernhagen J, Noels H. Diversity and Inter-Connections in the CXCR4 Chemokine Receptor/Ligand Family: Molecular Perspectives. Front Immunol 2015; 6:429. [PMID: 26347749 PMCID: PMC4543903 DOI: 10.3389/fimmu.2015.00429] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 08/07/2015] [Indexed: 12/19/2022] Open
Abstract
CXCR4 and its ligand CXCL12 mediate the homing of progenitor cells in the bone marrow and their recruitment to sites of injury, as well as affect processes such as cell arrest, survival, and angiogenesis. CXCL12 was long thought to be the sole CXCR4 ligand, but more recently the atypical chemokine macrophage migration inhibitory factor (MIF) was identified as an alternative, non-cognate ligand for CXCR4 and shown to mediate chemotaxis and arrest of CXCR4-expressing T-cells. This has complicated the understanding of CXCR4-mediated signaling and associated biological processes. Compared to CXCL12/CXCR4-induced signaling, only few details are known on MIF/CXCR4-mediated signaling and it remains unclear to which extent MIF and CXCL12 reciprocally influence CXCR4 binding and signaling. Furthermore, the atypical chemokine receptor 3 (ACKR3) (previously CXCR7) has added to the complexity of CXCR4 signaling due to its ability to bind CXCL12 and MIF, and to evoke CXCL12- and MIF-triggered signaling independently of CXCR4. Also, extracellular ubiquitin (eUb) and the viral protein gp120 (HIV) have been reported as CXCR4 ligands, whereas viral chemokine vMIP-II (Herpesvirus) and human β3-defensin (HBD-3) have been identified as CXCR4 antagonists. This review will provide insight into the diversity and inter-connections in the CXCR4 receptor/ligand family. We will discuss signaling pathways initiated by binding of CXCL12 vs. MIF to CXCR4, elaborate on how ACKR3 affects CXCR4 signaling, and summarize biological functions of CXCR4 signaling mediated by CXCL12 or MIF. Also, we will discuss eUb and gp120 as alternative ligands for CXCR4, and describe vMIP-II and HBD-3 as antagonists for CXCR4. Detailed insight into biological effects of CXCR4 signaling und underlying mechanisms, including diversity of CXCR4 ligands and inter-connections with other (chemokine) receptors, is clinically important, as the CXCR4 antagonist AMD3100 has been approved as stem cell mobilizer in specific disease settings.
Collapse
Affiliation(s)
- Lukas Pawig
- Institute of Molecular Cardiovascular Research (IMCAR), RWTH Aachen University , Aachen , Germany
| | - Christina Klasen
- Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University , Aachen , Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich , Munich , Germany ; DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance , Munich , Germany ; Cardiovascular Research Institute Maastricht (CARIM), Maastricht University , Maastricht , Netherlands
| | - Jürgen Bernhagen
- Institute of Biochemistry and Molecular Cell Biology, RWTH Aachen University , Aachen , Germany ; August-Lenz-Stiftung, Institute for Cardiovascular Research, Ludwig-Maximilians-University Munich , Munich , Germany
| | - Heidi Noels
- Institute of Molecular Cardiovascular Research (IMCAR), RWTH Aachen University , Aachen , Germany
| |
Collapse
|
49
|
Role of chemokine receptors CXCR4 and CXCR7 for platelet function. Biochem Soc Trans 2015; 43:720-6. [PMID: 26551719 DOI: 10.1042/bst20150113] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Indexed: 02/07/2023]
Abstract
Platelet-derived SDF-1α (stromal cell derived factor-α) mediates inflammation, immune defence and repair mechanisms at site of tissue injury. This review summarizes the relative expression of CXC chemokine receptor 4 (CXCR4) and CXCR7 in platelets, their dynamic trafficking in presence of ligands like chemokine C-X-C-motif ligand 11 (CXCL11), CXCL12 and MIF (macrophage migration inhibitory factor); how these receptors differentially mediate the functional and survival response to the chemokines CXCL11, CXCL12 and MIF. We further elaborate and emphasize the prognostic significance of platelet surface expression of CXCR4-CXCR7 in the context of coronary artery disease (CAD). SDF-1α/CXCL12, CXCL11, MIF effects mediated through CXCR4 and CXCR7 may play a regulatory role at the site of vascular and tissue inflammation, immune defence and repair where activated platelets reach as forerunners and function as critical players.
Collapse
|
50
|
Lapointe TK, Basso L, Iftinca MC, Flynn R, Chapman K, Dietrich G, Vergnolle N, Altier C. TRPV1 sensitization mediates postinflammatory visceral pain following acute colitis. Am J Physiol Gastrointest Liver Physiol 2015; 309:G87-99. [PMID: 26021808 DOI: 10.1152/ajpgi.00421.2014] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 05/20/2015] [Indexed: 01/31/2023]
Abstract
Quiescent phases of inflammatory bowel disease (IBD) are often accompanied by chronic abdominal pain. Although the transient receptor potential vanilloid 1 (TRPV1) ion channel has been postulated as an important mediator of visceral hypersensitivity, its functional role in postinflammatory pain remains elusive. This study aimed at establishing the role of TRPV1 in the peripheral sensitization underlying chronic visceral pain in the context of colitis. Wild-type and TRPV1-deficient mice were separated into three groups (control, acute colitis, and recovery), and experimental colitis was induced by oral administration of dextran sulfate sodium (DSS). Recovery mice showed increased chemically and mechanically evoked visceral hypersensitivity 5 wk post-DSS discontinuation, at which point inflammation had completely resolved. Significant changes in nonevoked pain-related behaviors could also be observed in these animals, indicative of persistent discomfort. These behavioral changes correlated with elevated colonic levels of substance P (SP) and TRPV1 in recovery mice, thus leading to the hypothesis that SP could sensitize TRPV1 function. In vitro experiments revealed that prolonged exposure to SP could indeed sensitize capsaicin-evoked currents in both cultured neurons and TRPV1-transfected human embryonic kidney (HEK) cells, a mechanism that involved TRPV1 ubiquitination and subsequent accumulation at the plasma membrane. Importantly, although TRPV1-deficient animals experienced similar disease severity and pain as wild-type mice in the acute phase of colitis, TRPV1 deletion prevented the development of postinflammatory visceral hypersensitivity and pain-associated behaviors. Collectively, our results suggest that chronic exposure of colon-innervating primary afferents to SP could sensitize TRPV1 and thus participate in the establishment of persistent abdominal pain following acute inflammation.
Collapse
Affiliation(s)
- Tamia K Lapointe
- Department of Physiology and Pharmacology, Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
| | - Lilian Basso
- Institut National de la Santé et de la Recherche Medicale (INSERM), Toulouse, France; Le Centre National de la Recherche Scientifique (CNRS), Toulouse, France; and Université de Toulouse III Paul Sabatier, Centre de Physiopathologie de Toulouse Purpan (CPTP), Toulouse, France
| | - Mircea C Iftinca
- Department of Physiology and Pharmacology, Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
| | - Robyn Flynn
- Department of Physiology and Pharmacology, Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
| | - Kevin Chapman
- Department of Physiology and Pharmacology, Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
| | - Gilles Dietrich
- Institut National de la Santé et de la Recherche Medicale (INSERM), Toulouse, France; Le Centre National de la Recherche Scientifique (CNRS), Toulouse, France; and Université de Toulouse III Paul Sabatier, Centre de Physiopathologie de Toulouse Purpan (CPTP), Toulouse, France
| | - Nathalie Vergnolle
- Department of Physiology and Pharmacology, Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada; Institut National de la Santé et de la Recherche Medicale (INSERM), Toulouse, France; Le Centre National de la Recherche Scientifique (CNRS), Toulouse, France; and Université de Toulouse III Paul Sabatier, Centre de Physiopathologie de Toulouse Purpan (CPTP), Toulouse, France
| | - Christophe Altier
- Department of Physiology and Pharmacology, Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada;
| |
Collapse
|